1
|
Dalali S, Kaviani F, Mahdevar M, Oroujalian A, Peymani M. Analysing glycolysis-related genes reveals the prognostic and diagnostic relevance of IER3 and AGRN in colorectal cancer. Genes Genomics 2025; 47:509-520. [PMID: 40048143 DOI: 10.1007/s13258-025-01618-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 01/04/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is a significant global health issue, with early detection being critical to improving patient survival. Dysregulation of the glycolysis pathway plays a pivotal role in CRC progression, but specific gene-level mechanisms remain underexplored. OBJECTIVE This study aimed to investigate the role of glycolysis-related genes in CRC development and identify potential diagnostic and prognostic biomarkers. METHODS We utilized The Cancer Genome Atlas (TCGA) dataset to perform differential expression analysis of glycolysis-related genes in CRC. Protein-protein interaction (PPI) network analysis was conducted to identify central hub genes. The diagnostic potential of selected genes was evaluated using ROC curve analysis, while their expression levels were validated through RT-qPCR. RESULTS IER3 and AGRN were identified as significantly upregulated genes associated with reduced survival rates in CRC patients. PPI analysis revealed their roles as central hub genes within the glycolysis pathway. ROC curve analysis demonstrated their ability to distinguish CRC patients from healthy individuals. Validation through RT-qPCR confirmed their significant overexpression in CRC samples, highlighting their involvement in disease progression. CONCLUSION IER3 and AGRN are critical components of the glycolysis pathway, driving CRC development and progression while also showing potential as biomarkers for predicting outcomes, diagnosing CRC, and serving as treatment targets.
Collapse
Affiliation(s)
- Samaneh Dalali
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Fatemeh Kaviani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mohammad Mahdevar
- Genius Gene, Genetics and Biotechnology Company, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Andisheh Oroujalian
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| |
Collapse
|
2
|
Han Y, Ma H, Tang Z, Jin C. Knockdown of IER3 Promotes Osteogenic Differentiation of Human Mesenchymal Stem Cells. Biomedicines 2025; 13:947. [PMID: 40299640 PMCID: PMC12025315 DOI: 10.3390/biomedicines13040947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 05/01/2025] Open
Abstract
Background: The differentiation process of human mesenchymal stem cells (hMSCs) is regulated by a variety of chemical, physical, and biological factors. These factors activate distinct signaling pathways and transcriptional networks, thereby regulating the lineage-specific differentiation of hMSCs. Objective: This study aims to investigate the role of Immediate Early Response 3 (IER3) in the osteogenic differentiation of human mesenchymal stem cells (hMSCs) and explore the underlying regulatory mechanisms by which IER3 influences osteogenesis. Methods: The expression levels of IER3 and osteogenesis-related genes were quantified when hMSCs were subjected to in vitro osteogenic induction. Then, stable IER3-knockdown hMSCs were generated using IER3-targeted shRNA lentiviral vectors, and the impact of IER3 on osteogenic differentiation was evaluated through both in vitro cell induction and hMSCs subcutaneous implantation model of nude mice. Moreover, RNA-seq and functional inhibition assays were performed to elucidate the signaling pathway through which IER3 regulates the osteogenic differentiation of hMSCs. Results: IER3 expression was significantly downregulated during osteogenic differentiation. Knockdown of IER3 markedly upregulated the expression of ALP and RUNX2, enhancing the osteogenic differentiation capacity of hMSCs, both in vitro and in vivo. Mechanistic studies revealed that IER3 knockdown significantly increased phosphorylated ERK1/2 levels, activating the MAPK/ERK signaling pathway. Furthermore, inhibition of the MAPK/ERK signaling pathway reversed the enhanced osteogenic differentiation observed following IER3 knockdown. Conclusions: Knockdown of IER3 promotes osteogenic differentiation of hMSCs through regulation of the MAPK/ERK signaling pathway, indicating IER3 represents a potential therapeutic target for the treatment of osteoporosis and bone defect-related diseases.
Collapse
Affiliation(s)
| | | | - Zhihui Tang
- Second Clinical Division, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing 100081, China; (Y.H.); (H.M.)
| | - Chanyuan Jin
- Second Clinical Division, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases, Beijing 100081, China; (Y.H.); (H.M.)
| |
Collapse
|
3
|
Moon KZ, Rahman MH, Alam MJ, Hossain MA, Hwang S, Kang S, Moon S, Park MN, Ahn CH, Kim B. Unraveling the interplay between cardiovascular diseases and alcohol use disorder: A bioinformatics and network-based exploration of shared molecular pathways and key biomarkers validation via western blot analysis. Comput Biol Chem 2025; 115:108338. [PMID: 39778286 DOI: 10.1016/j.compbiolchem.2024.108338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/02/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025]
Abstract
Clinical observations indicate a pronounced exacerbation of Cardiovascular Diseases (CVDs) in individuals grappling with Alcohol Use Disorder (AUD), suggesting an intricate interplay between these maladies. Pinpointing shared risk factors for both conditions has proven elusive. To address this, we pioneered a sophisticated bioinformatics framework and network-based strategy to unearth genes exhibiting aberrant expression patterns in both AUD and CVDs. In heart tissue samples from patients battling both AUD and CVDs, our study identified 76 Differentially Expressed Genes (DEGs) further used for retrieving important Gene Ontology (GO) keywords and metabolic pathways, highlighting mechanisms like proinflammatory cascades, T-cell cytotoxicity, antigen processing and presentation. By using Protein-Protein Interaction (PPI) analysis, we were able to identify key hub proteins that have a significant impact on the pathophysiology of these illnesses. Several hub proteins were identified include PTGS2, VCAM1, CCL2, CXCL8, IL7R, among these only CDH1 was covered in 10 algorithms of cytoHubba plugin. Furthermore, we pinpointed several Transcription Factors (TFs), including SOD2, CXCL8, THBS2, GREM1, CCL2, and PTGS2, alongside potential microRNAs (miRNAs) such as hsa-mir-203a-3p, hsa-mir-23a-3p, hsa-mir-98-5p, and hsa-mir-7-5p, which exert critical regulatory control over gene expression… In vitro study investigates the effect of alcohol on E-cadherin (CDH1) expression in HepG2 and Hep3B cells, showing a significant decrease in expression following ethanol treatment. These findings suggest that alcohol exposure may disrupt cell adhesion, potentially contributing to cellular changes associated with cardiovascular diseases. Our innovative approach has unveiled distinctive biomarkers delineating the dynamic interplay between AUD and various cardiovascular conditions for future therapeutic exploration.
Collapse
Affiliation(s)
- Kamelia Zaman Moon
- Department of Computer Science and Engineering, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushita 7003, Bangladesh.
| | - Md Jahangir Alam
- Department of Computer Science and Engineering, Islamic University, Kushita 7003, Bangladesh
| | - Md Arju Hossain
- Department of Biochemistry and Biotechnology, Khwaja Yunus Ali University, Sirajganj 6751, Bangladesh
| | - Sungho Hwang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Seungjoon Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Chi-Hoon Ahn
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
4
|
Shen J, Wang Y, Liu Y, Lan J, Long S, Li Y, Chen D, Yu P, Zhao J, Wang Y, Wang S, Yang F. Behavioral Abnormalities, Cognitive Impairments, Synaptic Deficits, and Gene Replacement Therapy in a CRISPR Engineered Rat Model of 5p15.2 Deletion Associated With Cri du Chat Syndrome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415224. [PMID: 39965128 PMCID: PMC11984882 DOI: 10.1002/advs.202415224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/29/2025] [Indexed: 02/20/2025]
Abstract
The Cri du Chat Syndrome (CdCS), a devastating genetic disorder caused by a deletion on chromosome 5p, faces challenges in finding effective treatments and accurate animal models. Using CRISPR-Cas9, a novel CdCS rat model with a 2q22 deletion is developed, mirroring a common genetic alteration in CdCS patients. This model exhibits pronounced deficits in social behavior, cognition, and anxiety, accompanied by neuronal abnormalities and immune dysregulation in key brain regions such as the hippocampus and medial prefrontal cortex (mPFC). The immunostaining and RNA-seq analyses provide new insights into CdCS pathogenesis, revealing inflammatory and immune processes. Importantly, it is demonstrated that early gene replacement therapy with AAV-Ctnnd2 alleviates cognitive impairments in CdCS rats, highlighting the potential for early intervention. However, the effectiveness of this therapy is confined to the early developmental stages and does not fully restore all CdCS symptoms. The findings deepen the understanding of CdCS pathogenesis and suggest promising therapeutic directions.
Collapse
Affiliation(s)
- Jingjing Shen
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Yan Wang
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Yang Liu
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
| | - Junying Lan
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
- Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijing100070China
- Laboratory of Cognitive and Behavioral DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijing100069China
| | - Shuang Long
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Yingbo Li
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Di Chen
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Peng Yu
- Chinese Institutes for Medical ResearchCapital Medical UniversityBeijing100069China
| | - Jing Zhao
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Yongjun Wang
- China National Clinical Research Center for Neurological DiseasesBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
- Department of NeurologyBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
- Clinical Center for Precision Medicine in StrokeCapital Medical UniversityBeijing100070China
- Center of Excellence for Omics Research (CORe)Beijing Tiantan HospitalCapital Medical UniversityBeijing100070China
| | - Shali Wang
- Institute of NeuroscienceSchool of Basic MedicineChongqing Medical UniversityChongqing400016China
| | - Feng Yang
- Basic and Translational Medicine CenterChina National Clinical Research Center for Neurological DiseasesBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
- Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijing100070China
- Laboratory of Cognitive and Behavioral DisordersBeijing Institute of Brain DisordersCapital Medical UniversityBeijing100069China
| |
Collapse
|
5
|
Xu S, Akhatayeva Z, Liu J, Feng X, Yu Y, Badaoui B, Esmailizadeh A, Kantanen J, Amills M, Lenstra JA, Johansson AM, Coltman DW, Liu GE, Curik I, Orozco-terWengel P, Paiva SR, Zinovieva NA, Zhang L, Yang J, Liu Z, Wang Y, Yu Y, Li M. Genetic advancements and future directions in ruminant livestock breeding: from reference genomes to multiomics innovations. SCIENCE CHINA. LIFE SCIENCES 2025; 68:934-960. [PMID: 39609363 DOI: 10.1007/s11427-024-2744-4] [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: 07/19/2024] [Accepted: 09/24/2024] [Indexed: 11/30/2024]
Abstract
Ruminant livestock provide a rich source of products, such as meat, milk, and wool, and play a critical role in global food security and nutrition. Over the past few decades, genomic studies of ruminant livestock have provided valuable insights into their domestication and the genetic basis of economically important traits, facilitating the breeding of elite varieties. In this review, we summarize the main advancements for domestic ruminants in reference genome assemblies, population genomics, and the identification of functional genes or variants for phenotypic traits. These traits include meat and carcass quality, reproduction, milk production, feed efficiency, wool and cashmere yield, horn development, tail type, coat color, environmental adaptation, and disease resistance. Functional genomic research is entering a new era with the advancements of graphical pangenomics and telomere-to-telomere (T2T) gap-free genome assembly. These advancements promise to improve our understanding of domestication and the molecular mechanisms underlying economically important traits in ruminant livestock. Finally, we provide new perspectives and future directions for genomic research on ruminant genomes. We suggest how ever-increasing multiomics datasets will facilitate future studies and molecular breeding in livestock, including the potential to uncover novel genetic mechanisms underlying phenotypic traits, to enable more accurate genomic prediction models, and to accelerate genetic improvement programs.
Collapse
Affiliation(s)
- Songsong Xu
- Frontiers Science Center for Molecular Design Breeding (MOE); State Key Laboratory of Animal Biotech Breeding; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhanerke Akhatayeva
- Institute of Grassland Research, Chinese Academy of Agricultural Sciences, Hohhot, 010010, China
| | - Jiaxin Liu
- Frontiers Science Center for Molecular Design Breeding (MOE); State Key Laboratory of Animal Biotech Breeding; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xueyan Feng
- Frontiers Science Center for Molecular Design Breeding (MOE); State Key Laboratory of Animal Biotech Breeding; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yi Yu
- Frontiers Science Center for Molecular Design Breeding (MOE); State Key Laboratory of Animal Biotech Breeding; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Bouabid Badaoui
- Laboratory of Biodiversity, Ecology and Genome, Department of Biology, Faculty of Sciences Rabat, Mohammed V University, Rabat, 10106, Morocco
| | - Ali Esmailizadeh
- Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman, 76169-133, Iran
| | - Juha Kantanen
- Production Systems, Natural Resources Institute Finland (Luke), Jokioinen, FI-31600, Finland
| | - Marcel Amills
- Department of Animal Genetics, Center for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus de la Universitat Autónoma de Barcelona, Bellaterra, 08193, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autónoma de Barcelona, Bellaterra, 08193, Spain
| | - Johannes A Lenstra
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584, The Netherlands
| | - Anna M Johansson
- Department of Animal Breeding and Genetics, Faculty of Veterinary Medicine and Animal Science, Swedish University of Agricultural Sciences, Uppsala, 75007, Sweden
| | - David W Coltman
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
- Department of Biology, Western University, London, Ontario, N6A 5B7, Canada
| | - George E Liu
- Animal Genomics and Improvement Laboratory, BARC, USDA-ARS, Beltsville, MD, 20705, USA
| | - Ino Curik
- Department of Animal Science, Faculty of Agriculture, University of Zagreb, Zagreb, 10000, Croatia
- Institute of Animal Sciences, Hungarian University of Agriculture and Life Sciences (MATE), Kaposvár, 7400, Hungary
| | | | - Samuel R Paiva
- Embrapa Genetic Resources and Biotechnology, Laboratory of Animal Genetics, Brasília, Federal District, 70770917, Brazil
| | - Natalia A Zinovieva
- L.K. Ernst Federal Science Center for Animal Husbandry, Moscow Region, Podolsk, 142132, Russian Federation
| | - Linwei Zhang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Ji Yang
- Frontiers Science Center for Molecular Design Breeding (MOE); State Key Laboratory of Animal Biotech Breeding; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhihong Liu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Yachun Wang
- Frontiers Science Center for Molecular Design Breeding (MOE); State Key Laboratory of Animal Biotech Breeding; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ying Yu
- Frontiers Science Center for Molecular Design Breeding (MOE); State Key Laboratory of Animal Biotech Breeding; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Menghua Li
- Frontiers Science Center for Molecular Design Breeding (MOE); State Key Laboratory of Animal Biotech Breeding; College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
- Sanya Institute of China Agricultural University, Sanya, 572024, China.
| |
Collapse
|
6
|
Kanduri M, Subhash S, Putino R, Mahale S, Kanduri C. IER3: exploring its dual function as an oncogene and tumor suppressor. Cancer Gene Ther 2025; 32:450-463. [PMID: 40090972 PMCID: PMC11976266 DOI: 10.1038/s41417-025-00891-y] [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: 11/01/2024] [Revised: 02/10/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
The IER3 gene has a complex role in cancer biology, acting either as a tumor suppressor or an oncogene, depending on the cancer type. This duality underscores the complexity and importance of molecular pathways in modulating cancer behavior. Despite its significance in cancer development, there is a dearth of studies elucidating the exact mechanisms underlying IER3's involvement in modulating cancer behavior. Here, utilizing cervical carcinoma and neuroblastoma (NB) cell lines as model systems we characterized the pathways that mediate the functional switch between the oncogenic and tumor suppressor roles of IER3. In HeLa cells, IER3 expression promotes an oncogenic program that includes immediate early response pathway genes such as EGR2, FOS, and JUN. However, in NB cells, IER3 suppresses the EGR2-dependent oncogenic program. This differential regulation of EGR2 by IER3 involves epigenetic modulation of the EGR2 promoter. IER3 dependent tumor suppressor pathway in NB cells relies on ADAM19 gene. Thus, our findings uncover the molecular pathways that dictate the context-dependent roles of IER3 in cancer, providing insights into its dual functionality in different cancer types.
Collapse
Affiliation(s)
- Meena Kanduri
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Santhilal Subhash
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Department of Biosciences and Bioengineering, Indian Institute of Technology Jammu, Jammu, India
| | - Rossana Putino
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sagar Mahale
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Chandrasekhar Kanduri
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
7
|
Zhao X, Zhang Y, Jia H, Lv L, Ahsan M, Fu X, Hu R, Shen Z, Shen N. Diversities of African swine fever virus host-virus dynamics revealed by single-cell profiling. J Virol 2025; 99:e0203524. [PMID: 39932318 PMCID: PMC11917525 DOI: 10.1128/jvi.02035-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/08/2025] [Indexed: 03/19/2025] Open
Abstract
African swine fever virus (ASFV) causes epidemics with high mortality; however, effective vaccines and therapies remain missing. Here, we depict a temporal single-cell landscape of primary porcine alveolar macrophages (PAMs) exposed to three different virulent ASFV strains in vitro. We found that attenuated and low-virulence ASFV strains tend to exhibit higher viral loads than highly virulent strain, which may result from upregulated RNA polymerase subunit genes expression. On the host side, our study highlights the IRF7-mediated positive feedback loop to the activation of the interferon signaling pathway in cells exposed to attenuated and low virulent ASFV strains. Moreover, we unraveled the PAMs populations marked by expressions of the IFI16 and CD163, respectively, which produce high levels of interferon-stimulated genes (ISGs) and IL18 to regulate the host response to different virulent ASFV strains. Collectively, our data provide insights into the complex host-virus interactions with various ASFV strain infections, which may shed light on the development of effective antiviral strategies.IMPORTANCEThere is still no available research on the temporal transcriptional profile of host cells exposed to different virulent ASFV strains at the single-cell level. Here, we first profiled the temporal viral and host transcriptomes in PAMs exposed to high virulent, attenuated virulent, and low virulent ASFV strains. Our analysis revealed that attenuated and low-virulence ASFV strains tend to exhibit higher viral loads than highly virulent strains, which may result from upregulated RNA polymerase subunit genes expression. We also found a positive feedback loop of the interferon signaling pathway mediated through IRF7 and identified the populations of PAMs marked by IFI6 and CD163, respectively, which produce high levels of ISGs and IL18 to regulate host response to different virulent ASFV strains. Our study delineated a comprehensive single-cell landscape of host-virus dynamics across ASFV strains with different virulences and would provide an important resource for future research.
Collapse
MESH Headings
- African Swine Fever Virus/pathogenicity
- African Swine Fever Virus/physiology
- African Swine Fever Virus/genetics
- Animals
- Swine
- Single-Cell Analysis
- African Swine Fever/virology
- African Swine Fever/immunology
- African Swine Fever/genetics
- Macrophages, Alveolar/virology
- Macrophages, Alveolar/metabolism
- Macrophages, Alveolar/immunology
- Host-Pathogen Interactions
- Interferon Regulatory Factor-7/metabolism
- Interferon Regulatory Factor-7/genetics
- Viral Load
- CD163 Antigen
- Signal Transduction
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Interferons/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Virulence
Collapse
Affiliation(s)
- Xiaoyang Zhao
- Department of
Obstetrics and Gynecology of Sir Run Run Shaw Hospital & Liangzhu
Laboratory, Zhejiang University School of
Medicine, Hangzhou,
China
| | - Yanyan Zhang
- Changchun Veterinary
Research Institute, Chinese Academy of Agricultural
Sciences, Changchun,
China
| | - Hanying Jia
- Liangzhu Laboratory,
Zhejiang University School of Medicine, Hangzhou,
China
| | - Lin Lv
- Department of
Obstetrics and Gynecology of Sir Run Run Shaw Hospital & Liangzhu
Laboratory, Zhejiang University School of
Medicine, Hangzhou,
China
| | - Md.Asif Ahsan
- Department of
Obstetrics and Gynecology of Sir Run Run Shaw Hospital & Liangzhu
Laboratory, Zhejiang University School of
Medicine, Hangzhou,
China
| | - Xudong Fu
- Department of
Obstetrics and Gynecology of Sir Run Run Shaw Hospital & Liangzhu
Laboratory, Zhejiang University School of
Medicine, Hangzhou,
China
| | - Rongliang Hu
- Changchun Veterinary
Research Institute, Chinese Academy of Agricultural
Sciences, Changchun,
China
| | - Zhiqiang Shen
- Shandong Lvdu
Bio-Sciences and Technology Co., Ltd.,
Binzhou, Shandong,
China
- Shandong Binzhou
Academy of Animal Science and Veterinary Medicine, Shandong Academy of
Agricultural Sciences, Binzhou,
Shandong, China
| | - Ning Shen
- Department of
Obstetrics and Gynecology of Sir Run Run Shaw Hospital & Liangzhu
Laboratory, Zhejiang University School of
Medicine, Hangzhou,
China
| |
Collapse
|
8
|
Mezzetti M, Zontini AM, Minuti A, Yoon I, Trevisi E. Impact of a Saccharomyces cerevisiae Fermentation Product Supplemented from 20 Days Before Dry-Off Through 60 Days of Lactation on the Metabolic Adaptation of Dairy Cows to the Peripartum Phase. Animals (Basel) 2025; 15:480. [PMID: 40002962 PMCID: PMC11851521 DOI: 10.3390/ani15040480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/04/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Sixty Holstein cows were enrolled at -76 days from calving (DFC) and classified based on the daily SCC during the previous week from an automated milking system. The separation thresholds for low (L, n = 46) and high (H, n = 14) classifications were 100 K/mL for primiparous and 200 K/mL for multiparous cows. Cows were then assigned to two homogeneous groups to receive diets supplemented with 19 g/d of a Saccharomyces cerevisiae fermentation product (TRT; NutriTek, Diamond V, Cedar Rapids, IA, USA) or without supplementation (CTR) until 60 DFC. Cows were dried off at -56 DFC and monitored for disease incidence, milk yield and composition, plasma metabolic profile, and whole blood count from -76 to 60 DFC. Data were analyzed utilizing ANOVA and mixed models for repeated measures. During the dry period, TRT cows had greater plasma thiol and albumin compared to CTR. TRT-L cows had greater plasma protein and globulin than CTR-L. TRT-H cows had heightened hematocrit; reduced plasma globulin and haptoglobin; and higher albumin, albumin to globulin ratio, and thiol than CTR-H. TRT-H cows had greater concentrations of leukocytes and lymphocytes and lower plasma protein and ceruloplasmin at -54 DFC; lower reactive oxygen species to ferric ion-reducing antioxidant power ratios at -44 DFC; and greater concentrations of lymphocytes and plasma gamma glutamyl transferase at -7 DFC than CTR-H. After calving, TRT cows had a lower incidence of mastitis and higher butterfat, as well as greater plasma haptoglobin and aspartate amino transferase (AST) and reduced Mg compared to CTR. TRT cows had lower SCC between 1 and 7 DFC and a greater ECM between 41 and 60 DFC compared to CTR. TRT-H cows had lower SCC between 1 and 7 DFC and greater hemoglobin and plasma AST than CTR-H. Ameliorated immune system functions due to Saccharomyces cerevisiae fermentation product administration lowered the SCC in TRT-H cows and prevented the onset of new intramammary infections across both L and H SCC groups, supporting the improved productive performance of dairy cows.
Collapse
Affiliation(s)
- Matteo Mezzetti
- Department of Animal Sciences, Food and Nutrition (DIANA), Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, 29122 Piacenza, PC, Italy; (M.M.); (A.M.)
| | | | - Andrea Minuti
- Department of Animal Sciences, Food and Nutrition (DIANA), Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, 29122 Piacenza, PC, Italy; (M.M.); (A.M.)
| | | | - Erminio Trevisi
- Department of Animal Sciences, Food and Nutrition (DIANA), Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, 29122 Piacenza, PC, Italy; (M.M.); (A.M.)
| |
Collapse
|
9
|
Kaya VO, Adebali O. UV-induced reorganization of 3D genome mediates DNA damage response. Nat Commun 2025; 16:1376. [PMID: 39910043 PMCID: PMC11799157 DOI: 10.1038/s41467-024-55724-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 12/20/2024] [Indexed: 02/07/2025] Open
Abstract
While it is well-established that UV radiation threatens genomic integrity, the precise mechanisms by which cells orchestrate DNA damage response and repair within the context of 3D genome architecture remain unclear. Here, we address this gap by investigating the UV-induced reorganization of the 3D genome and its critical role in mediating damage response. Employing temporal maps of contact matrices and transcriptional profiles, we illustrate the immediate and holistic changes in genome architecture post-irradiation, emphasizing the significance of this reconfiguration for effective DNA repair processes. We demonstrate that UV radiation triggers a comprehensive restructuring of the 3D genome organization at all levels, including loops, topologically associating domains and compartments. Through the analysis of DNA damage and excision repair maps, we uncover a correlation between genome folding, gene regulation, damage formation probability, and repair efficacy. We show that adaptive reorganization of the 3D genome is a key mediator of the damage response, providing new insights into the complex interplay of genomic structure and cellular defense mechanisms against UV-induced damage, thereby advancing our understanding of cellular resilience.
Collapse
Affiliation(s)
- Veysel Oğulcan Kaya
- Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, Türkiye
| | - Ogün Adebali
- Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, Türkiye.
| |
Collapse
|
10
|
Ding H, Shi X, Ma J, Cao C, Liu Y, Lu J, Bai L, Li X, Li H. Integrative transcriptomic analysis reveals Cd72 as a novel pro-inflammatory factor in microglia following experimental ischemic stroke. Exp Neurol 2024; 382:114974. [PMID: 39326825 DOI: 10.1016/j.expneurol.2024.114974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/07/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
Ischemic stroke remains a leading cause of global mortality and disability, with neuroinflammation playing a critical role in determining patient outcomes. Microglia, the brain's resident immune cells, can both exacerbate neuroinflammation and neuronal damage by releasing neurotoxic mediators and engaging in excessive phagocytosis, while also aiding recovery through the production of anti-inflammatory cytokines and debris clearance. However, the molecular mechanisms governing microglial activation and polarization after ischemic stroke are not well elucidated. In this study, we combined integrative transcriptomic analyses with experimental validation in a murine model of middle cerebral artery occlusion/reperfusion (MCAO/R) to explore microglial heterogeneity and identify key regulatory factors in ischemic stroke. Bioinformatics analysis identified Cd72 as a novel pro-inflammatory modulator within ischemia-associated microglial phenotypes. We observed significant upregulation of Cd72 in microglia following MCAO/R, and selective knockdown of Cd72 using CX3CR1Cre/ERT2 mice and Cre recombinase-dependent adeno-associated virus reduced MCAO/R-induced infarct volume, neuronal apoptosis, and neurological deficits. Furthermore, Cd72 expression in microglia was positively correlated with pro-inflammatory pathways and cytokines, including TNF-α, IL-1β, and IL-6. Knockdown of Cd72 significantly reduced these pro-inflammatory factors, highlighting its potential as a therapeutic target for mitigating inflammation in ischemic stroke. In conclusion, this study identifies Cd72 as a critical pro-inflammatory regulator in microglia following ischemic stroke, with its knockdown effectively reducing neuroinflammation and associated brain injury, highlighting Cd72 as a promising therapeutic target.
Collapse
Affiliation(s)
- Haojie Ding
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xuan Shi
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Junwei Ma
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Chang Cao
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Yangyang Liu
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Jinxin Lu
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Lei Bai
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xiang Li
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| | - Haiying Li
- Department of Neurosurgery, Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| |
Collapse
|
11
|
Bedoux A, Lallemant-Dudek P, Bensidhoum M, Potier E, Larochette N, Mary P, Vialle R, Hoc T, Bachy M. Effects of shear stress on mesenchymal stem cells of patients with osteogenesis imperfecta. Orthop Traumatol Surg Res 2024:104067. [PMID: 39581491 DOI: 10.1016/j.otsr.2024.104067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 03/15/2024] [Accepted: 04/04/2024] [Indexed: 11/26/2024]
Abstract
INTRODUCTION Osteogenesis imperfecta (OI) is a rare genetic bone disorder, mainly caused by autosomal dominant mutations of the COL1A1 or COL1A2 genes that encode the alpha chains of type 1 collagen. In severe forms and in nonambulatory patients, for whom physical exercise is difficult, exposing the bone to mechanical stimuli by promoting movement, especially with physiotherapy and mobility aids, is an essential part of clinical practice. However, the effects of mechanical stimulation at the cellular level remain unknown for this disease. HYPOTHESIS The study hypothesis was that human mesenchymal stem cells (hMSCs) from patients with OI were as sensitive to mechanical stimulation as those from healthy patients, validating the current clinical practice. MATERIALS AND METHODS hMSCs were harvested from 3 healthy control subjects and 3 patients with OI during an elective osteotomy of a long bone of the lower limb. The healthy and OI hMSCs were then exposed to mechanical stimuli, such as intermittent shear stress of 0, 0.7, 1.5, and 3 Pascal (Pa) at a frequency of 2.8 Hertz (Hz) for 30 minutes using a commercial ibidi system. The immediate early gene expression of themechanosensitive prostaglandin-endoperoxide synthase 2 (PTGS2) was examined 1 hour after stimulation to determine the best level of mechanical stimulation. The expression of 7 other mechanosensitive genes was also examined for this level of mechanical stimulation after applying intermittent shear stress at 1.5 Pa. RESULTS In all hMSCs, mechanical stimulation induced PTGS2 gene overexpression with a maximum after exposure to intermittent shear stress of 1.5 Pa and without significant differences between OI and healthy donors. Except for fibroblast growth factor 2, gene expression in OI donors was found to be significantly different from that in hMSCs not exposed to shear stress. Moreover, the relative expression associated with mechanical stimulation was not significantly different between healthy and OI donors for most other genes. DISCUSSION This is the first study to demonstrate that hMSCs from patients with OI are as sensitive to mechanical shear stress as those from healthy donors. The mechanical stress that resulted in the greatest change in the expression of PTGS2 in patients with OI was similar to that previously reported in the literature for healthy subjects. These findings are an important step toward further fundamental research aimed at confirming the effects of mechanical stress at the cellular level over the long term and, more importantly, toward developing clinical protocols for delivering mechanical stimuli to these patients. LEVEL OF EVIDENCE III; comparative case-control study.
Collapse
Affiliation(s)
- Agathe Bedoux
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France
| | - Pauline Lallemant-Dudek
- Service de Médecine Physique et Réadaptation fonctionnelle pédiatrique, APHP-Sorbonne Université, Hôpital Armand Trousseau, 26 avenue du Docteur Arnold-Netter, 75012 Paris, France
| | - Morad Bensidhoum
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France
| | - Esther Potier
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France
| | - Nathanael Larochette
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France
| | - Pierre Mary
- Service de Chirurgie Orthopédique et Réparatrice de l'Enfant, APHP-Sorbonne Université, Hôpital Armand Trousseau, 26 avenue du Docteur Arnold-Netter, 75012 Paris, France
| | - Raphaël Vialle
- Service de Chirurgie Orthopédique et Réparatrice de l'Enfant, APHP-Sorbonne Université, Hôpital Armand Trousseau, 26 avenue du Docteur Arnold-Netter, 75012 Paris, France
| | - Thierry Hoc
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France; Ecole Centrale de Lyon, Département de mécanique, MSGMGC, 36 Avenue Guy de Collongue, 69134 Ecully Cedex, France
| | - Manon Bachy
- Université de Paris Cité, B3OA, UMR CNRS 7052, INSERM U1271, 10 avenue de Verdun, 75010 Paris, France; Service de Chirurgie Orthopédique et Réparatrice de l'Enfant, APHP-Sorbonne Université, Hôpital Armand Trousseau, 26 avenue du Docteur Arnold-Netter, 75012 Paris, France.
| |
Collapse
|
12
|
Küppers O, Ahmad M, Haffner-Luntzer M, Scharffetter-Kochanek K, Ignatius A, Fischer V. Inflammatory priming of human mesenchymal stem cells induces osteogenic differentiation via the early response gene IER3. FASEB J 2024; 38:e70076. [PMID: 39373973 DOI: 10.1096/fj.202401344r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/27/2024] [Accepted: 09/16/2024] [Indexed: 10/08/2024]
Abstract
Mesenchymal stem cells (MSCs) have gained tremendous interest due to their overall potent pro-regenerative and immunomodulatory properties. In recent years, various in vitro and preclinical studies have investigated different priming ("licensing") approaches to enhance MSC functions for specific therapeutic purposes. In this study, we primed bone marrow-derived human MSCs (hMSCs) with an inflammation cocktail designed to mimic the elevated levels of inflammatory mediators found in serum of patients with severe injuries, such as bone fractures. We observed a significantly enhanced osteogenic differentiation potential of primed hMSCs compared to untreated controls. By RNA-sequencing analysis, we identified the immediate early response 3 (IER3) gene as one of the top-regulated genes upon inflammatory priming. Small interfering RNA knockdown experiments established IER3 as a novel positive regulator of osteogenic differentiation. Mechanistic analysis further revealed that IER3 deletion significantly downregulated bone marrow stromal cell antigen 2 (BST2) expression and extracellular signal-related kinase 1/2 (ERK1/2) phosphorylation in hMSCs, suggesting that IER3 regulates osteogenic differentiation through BST2 and ERK1/2 signaling pathway activation. On the basis of these findings, we propose IER3 as a novel therapeutic target to promote hMSC osteoblastogenesis, which might be of high clinical relevance, for example, in patients with osteoporosis or compromised fracture healing.
Collapse
Affiliation(s)
- Oliver Küppers
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Mubashir Ahmad
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | | | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
13
|
McFarlin BK, Bridgeman EA, Curtis JH, Vingren JL, Hill DW. Baker's yeast beta glucan supplementation was associated with an improved innate immune mRNA expression response after exercise. Methods 2024; 230:68-79. [PMID: 39097177 DOI: 10.1016/j.ymeth.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024] Open
Abstract
Beta glucans are found in many natural sources, however, only Baker's Yeast Beta Glucan (BYBG) has been well documented to have structure-function effects that are associated with improved innate immune response to stressors (e.g., exercise, infection, etc.). The purpose was to identify a BYBG-associated mRNA expression pattern following exercise. Participants gave IRB-approved consent and were randomized to BYBG (Wellmune®; N=9) or Placebo (maltodextrin; N=10) for 6-weeks prior to performing 90 min of whole-body exercise. Paxgene blood samples were collected prior to exercise (PRE), after exercise (POST), two hours after exercise (2H), and four hours after exercise (4H). Total RNA was isolated and analyzed for the expression of 770 innate immune response mRNA (730 mRNA targets; 40 housekeepers/controls; Nanostring nCounter). The raw data were normalized against housekeeping controls and expressed as Log2 fold change from PRE for a given condition. Significance was set at p < 0.05 with adjustments for multiple comparisons and false discovery rate. We identified 47 mRNA whose expression was changed after exercise with BYBG and classified them to four functional pathways: 1) Immune Cell Maturation (8 mRNA), 2) Immune Response and Function (5 mRNA), 3) Pattern Recognition Receptors and DAMP or PAMP Detection (25 mRNA), and 4) Detection and Resolution of Tissue Damage (9 mRNA). The identified mRNA whose expression was altered after exercise with BYBG may represent an innate immune response pattern and supports previous conclusions that BYBG improves immune response to a future sterile inflammation or infection.
Collapse
Affiliation(s)
- Brian K McFarlin
- Applied Physiology Laboratory, University of North Texas, Denton, TX 76203, United States; University of North Texas, Dept. of Biological Sciences, Denton, TX 76203, United States.
| | - Elizabeth A Bridgeman
- Applied Physiology Laboratory, University of North Texas, Denton, TX 76203, United States.
| | - John H Curtis
- Applied Physiology Laboratory, University of North Texas, Denton, TX 76203, United States.
| | - Jakob L Vingren
- Applied Physiology Laboratory, University of North Texas, Denton, TX 76203, United States; University of North Texas, Dept. of Biological Sciences, Denton, TX 76203, United States.
| | - David W Hill
- Applied Physiology Laboratory, University of North Texas, Denton, TX 76203, United States.
| |
Collapse
|
14
|
Wang Q, Zhang C, Pang Y, Cheng M, Wang R, Chen X, Ji T, Yang Y, Zhang J, Zhong C. Comprehensive analysis of bulk, single-cell RNA sequencing, and spatial transcriptomics revealed IER3 for predicting malignant progression and immunotherapy efficacy in glioma. Cancer Cell Int 2024; 24:332. [PMID: 39354533 PMCID: PMC11443732 DOI: 10.1186/s12935-024-03511-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND As part of stress-triggered molecules, immediate early response 3 (IER3) dysregulation has been reported to sustain pro-oncogenic pathways and precede malignant transformation. However, the role of IER3 in glioma pathology is ill-defined. METHODS Immunohistochemistry (IHC) assay and publicly available glioma datasets were used to calculate the IER3 expression level in glioma. Wound healing, invasion and cell counting kit-8 (CCK8) assays were applied to measure the cell viability and capacities of migration and invasion of glioma cells in vitro. The immunofluorescence (IF) assay was used to assess the expression associations of IER3 with CCL2 and TGFBI. Cox regression analysis and Kaplan-Meier (K-M) curve were introduced to compute the prognosis-predicting value of IER3. Variations in copy number (CNVs), single nucleotide (SNVs), and methylation profiles were analyzed to illustrate the epigenetic modifications of IER3. Gliomas were divided into two subgroups using the restricted cubic spline (RCS) method. RESULTS IER3: was overexpressed and hypomethylated in gliomas and significantly associated with the dismal prognosis of glioma samples. Samples in the high IER3 subgroup were characterized by increased infiltration of tumor-associated monocytes/macrophages (TAMMs), as well as the elevated sensitivity to Dabrafenib, an inhibitor of BRAF. In addition, this subgroup demonstrated a low mutation rate of IDH, high gain rates of BRAF, ELTD1, and PDGFA. Gliomas with relatively low IER3 expression demonstrated a less invasive subtype and were featured by favorable prognosis, increased response to immunotherapy, and adjuvant chemotherapy plus radiotherapy. The IF assay revealed that IER3 was co-localized and co-expressed with TGFBI. The glioma cells with small interfering RNA (siRNA)-silenced IER3 displayed lower migration, invasion, proliferation, and cell viability than the control group. CONCLUSIONS In this study, we identified IER3 upregulation as an essential biomarker that could assist in adjuvant therapy and prognosis prediction for gliomas.
Collapse
Affiliation(s)
- Qi Wang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunyu Zhang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ying Pang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Meng Cheng
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rui Wang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xu Chen
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tongjie Ji
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuntong Yang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing Zhang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute for Advanced Study, Tongji University, Shanghai, China
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
15
|
Cui D, Yu T. Unveiling the glycolysis in sepsis: Integrated bioinformatics and machine learning analysis identifies crucial roles for IER3, DSC2, and PPARG in disease pathogenesis. Medicine (Baltimore) 2024; 103:e39867. [PMID: 39331858 PMCID: PMC11441936 DOI: 10.1097/md.0000000000039867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 08/26/2024] [Indexed: 09/29/2024] Open
Abstract
Sepsis, a multifaceted syndrome driven by an imbalanced host response to infection, remains a significant medical challenge. At its core lies the pivotal role of glycolysis, orchestrating immune responses especially in severe sepsis. The intertwined dynamics between glycolysis, sepsis, and immunity, however, have gaps in knowledge with several Crucial genes still shrouded in ambiguity. We harvested transcriptomic profiles from the peripheral blood of 107 septic patients juxtaposed against 29 healthy controls. Delving into this dataset, differential expression analysis shed light on genes distinctly linked to glycolysis in both cohorts. Harnessing the prowess of LASSO regression and SVM-RFE, we isolated Crucial genes, paving the way for a sepsis risk prediction model, subsequently vetted via Calibration and decision curve analysis. Using the CIBERSORT algorithm, we further mapped 22 immune cell subtypes within the septic samples, establishing potential interactions with the delineated Crucial genes. Our efforts unveiled 21 genes intricately tied to glycolysis that exhibited differential expression patterns. Gene set enrichment analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses offered insights, spotlighting pathways predominantly associated with oxidative phosphorylation, PPAR signaling pathway, Glycolysis/Gluconeogenesis and HIF-1 signaling pathway. Among the myriad genes, IER3, DSC2, and PPARG emerged as linchpins, their prominence in sepsis further validated through ROC analytics. These sentinel genes demonstrated profound affiliations with various immune cell facets, bridging the complex terrain of glycolysis, sepsis, and immune responses. In line with our endeavor to "unveil the glycolysis in sepsis," the discovery of IER3, DSC2, and PPARG reinforces their cardinal roles in sepsis pathogenesis. These revelations accentuate the intricate dance between glycolysis and immunological shifts in septic conditions, offering novel avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Dongqing Cui
- Emergency Department, Beijing Sixth Hospital, Beijing, China
| | - Tian Yu
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
McQueen LW, Ladak SS, Layton GR, Wozniak M, Solomon C, El-Dean Z, Murphy GJ, Zakkar M. Spatial Transcriptomic Profiling of Human Saphenous Vein Exposed to Ex Vivo Arterial Haemodynamics-Implications for Coronary Artery Bypass Graft Patency and Vein Graft Disease. Int J Mol Sci 2024; 25:10368. [PMID: 39408698 PMCID: PMC11476946 DOI: 10.3390/ijms251910368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
Vein graft disease is the process by which saphenous vein grafts, utilised for revascularisation during coronary artery bypass graft surgery, undergo an inflammation-driven intimal hyperplasia and accelerated atherosclerosis process in subsequent years after implantation. The role of the arterial circulation, particularly the haemodynamic properties' impact on graft patency, have been investigated but have not to date been explored in depth at the transcriptomic level. We have undertaken the first-in-man spatial transcriptomic analysis of the long saphenous vein in response to ex vivo acute arterial haemodynamic stimulation, utilising a combination of a custom 3D-printed perfusion bioreactor and the 10X Genomics Visium Spatial Gene Expression technology. We identify a total of 413 significant genes (372 upregulated and 41 downregulated) differentially expressed in response to arterial haemodynamic conditions. These genes were associated with pathways including NFkB, TNF, MAPK, and PI3K/Akt, among others. These are established pathways involved in the initiation of an early pro-inflammatory response, leukocyte activation and adhesion signalling, tissue remodelling, and cellular differentiation. Utilising unsupervised clustering analysis, we have been able to classify subsets of the expression based on cell type and with spatial resolution. These findings allow for further characterisation of the early saphenous vein graft transcriptional landscape during the earliest stage of implantation that contributes to vein graft disease, in particular validation of pathways and druggable targets that could contribute towards the therapeutic inhibition of processes underpinning vein graft disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mustafa Zakkar
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE1 7RH, UK; (L.W.M.); (S.S.L.); (G.R.L.); (M.W.); (C.S.); (Z.E.-D.); (G.J.M.)
| |
Collapse
|
17
|
Li J, Yang G, Liu J, Li G, Zhou H, He Y, Fei X, Zhao D. Integrating transcriptomics, eQTL, and Mendelian randomization to dissect monocyte roles in severe COVID-19 and gout flare. Front Genet 2024; 15:1385316. [PMID: 39385934 PMCID: PMC11461236 DOI: 10.3389/fgene.2024.1385316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction There are considerable similarities between the pathophysiology of gout flare and the dysregulated inflammatory response in severe COVID-19 infection. Monocytes are the key immune cells involved in the pathogenesis of both diseases. Therefore, it is critical to elucidate the molecular basis of the function of monocytes in gout and COVID-19 in order to develop more effective therapeutic approaches. Methods The single-cell RNA sequencing (scRNA-seq), large-scale genome-wide association studies (GWAS), and expression quantitative trait loci (eQTL) data of gout and severe COVID-19 were comprehensively analyzed. Cellular heterogeneity and intercellular communication were identified using the scRNA-seq datasets, and the monocyte-specific differentially expressed genes (DEGs) between COVID-19, gout and normal subjects were screened. In addition, the correlation of the DEGs with severe COVID-19 and gout flare was analyzed through GWAS statistics and eQTL data. Results The scRNA-seq analysis exhibited that the proportion of classical monocytes was increased in both severe COVID-19 and gout patient groups compared to healthy controls. Differential expression analysis and MR analysis showed that NLRP3 was positively associated with the risk of severe COVID-19 and involved 11 SNPs, of which rs4925547 was not significantly co-localized. In contrast, IER3 was positively associated with the risk of gout and involved 9 SNPs, of which rs1264372 was significantly co-localized. Discussion Monocytes have a complex role in gout flare and severe COVID-19, which underscores the potential mechanisms and clinical significance of the interaction between the two diseases.
Collapse
Affiliation(s)
- Jiajia Li
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Guixian Yang
- Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Junnan Liu
- Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Guofeng Li
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Huiling Zhou
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yuan He
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xinru Fei
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Dongkai Zhao
- Third Affiliated Clinical Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
18
|
Graham MK, Wang R, Chikarmane R, Abel B, Vaghasia A, Gupta A, Zheng Q, Hicks J, Sysa-Shah P, Pan X, Castagna N, Liu J, Meyers J, Skaist A, Zhang Y, Rubenstein M, Schuebel K, Simons BW, Bieberich CJ, Nelson WG, Lupold SE, DeWeese TL, De Marzo AM, Yegnasubramanian S. Convergent alterations in the tumor microenvironment of MYC-driven human and murine prostate cancer. Nat Commun 2024; 15:7414. [PMID: 39198404 PMCID: PMC11358296 DOI: 10.1038/s41467-024-51450-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
How prostate cancer cells and their precursors mediate changes in the tumor microenvironment (TME) to drive prostate cancer progression is unclear, in part due to the inability to longitudinally study the disease evolution in human tissues. To overcome this limitation, we perform extensive single-cell RNA-sequencing (scRNA-seq) and molecular pathology of the comparative biology between human prostate cancer and key stages in the disease evolution of a genetically engineered mouse model (GEMM) of prostate cancer. Our studies of human tissues reveal that cancer cell-intrinsic activation of MYC signaling is a common denominator across the well-known molecular and pathological heterogeneity of human prostate cancer. Cell communication network and pathway analyses in GEMMs show that MYC oncogene-expressing neoplastic cells, directly and indirectly, reprogram the TME during carcinogenesis, leading to a convergence of cell state alterations in neighboring epithelial, immune, and fibroblast cell types that parallel key findings in human prostate cancer.
Collapse
Affiliation(s)
- Mindy K Graham
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Urology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Rulin Wang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Roshan Chikarmane
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Bulouere Abel
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Ajay Vaghasia
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Anuj Gupta
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Qizhi Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Jessica Hicks
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Polina Sysa-Shah
- The Brady Urological Institute and Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Xin Pan
- Department of Neurology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Nicole Castagna
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Jianyong Liu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Jennifer Meyers
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Alyza Skaist
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Yan Zhang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Michael Rubenstein
- Department of Biological Sciences, University of Maryland at Baltimore County, Baltimore, MD, USA
| | - Kornel Schuebel
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Brian W Simons
- Center for Comparative Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Charles J Bieberich
- Department of Biological Sciences, University of Maryland at Baltimore County, Baltimore, MD, USA
| | - William G Nelson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- The Brady Urological Institute and Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Shawn E Lupold
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- The Brady Urological Institute and Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Theodore L DeWeese
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- The Brady Urological Institute and Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Angelo M De Marzo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- The Brady Urological Institute and Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Srinivasan Yegnasubramanian
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
- inHealth Precision Medicine Program, Johns Hopkins Medicine, Baltimore, MD, USA.
| |
Collapse
|
19
|
Gatti DM, Tyler AL, Mahoney JM, Churchill GA, Yener B, Koyuncu D, Gurcan MN, Niazi MKK, Tavolara T, Gower A, Dayao D, McGlone E, Ginese ML, Specht A, Alsharaydeh A, Tessier PA, Kurtz SL, Elkins KL, Kramnik I, Beamer G. Systems genetics uncover new loci containing functional gene candidates in Mycobacterium tuberculosis-infected Diversity Outbred mice. PLoS Pathog 2024; 20:e1011915. [PMID: 38861581 PMCID: PMC11195971 DOI: 10.1371/journal.ppat.1011915] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/24/2024] [Accepted: 04/17/2024] [Indexed: 06/13/2024] Open
Abstract
Mycobacterium tuberculosis infects two billion people across the globe, and results in 8-9 million new tuberculosis (TB) cases and 1-1.5 million deaths each year. Most patients have no known genetic basis that predisposes them to disease. Here, we investigate the complex genetic basis of pulmonary TB by modelling human genetic diversity with the Diversity Outbred mouse population. When infected with M. tuberculosis, one-third develop early onset, rapidly progressive, necrotizing granulomas and succumb within 60 days. The remaining develop non-necrotizing granulomas and survive longer than 60 days. Genetic mapping using immune and inflammatory mediators; and clinical, microbiological, and granuloma correlates of disease identified five new loci on mouse chromosomes 1, 2, 4, 16; and three known loci on chromosomes 3 and 17. Further, multiple positively correlated traits shared loci on chromosomes 1, 16, and 17 and had similar patterns of allele effects, suggesting these loci contain critical genetic regulators of inflammatory responses to M. tuberculosis. To narrow the list of candidate genes, we used a machine learning strategy that integrated gene expression signatures from lungs of M. tuberculosis-infected Diversity Outbred mice with gene interaction networks to generate scores representing functional relationships. The scores were used to rank candidates for each mapped trait, resulting in 11 candidate genes: Ncf2, Fam20b, S100a8, S100a9, Itgb5, Fstl1, Zbtb20, Ddr1, Ier3, Vegfa, and Zfp318. Although all candidates have roles in infection, inflammation, cell migration, extracellular matrix remodeling, or intracellular signaling, and all contain single nucleotide polymorphisms (SNPs), SNPs in only four genes (S100a8, Itgb5, Fstl1, Zfp318) are predicted to have deleterious effects on protein functions. We performed methodological and candidate validations to (i) assess biological relevance of predicted allele effects by showing that Diversity Outbred mice carrying PWK/PhJ alleles at the H-2 locus on chromosome 17 QTL have shorter survival; (ii) confirm accuracy of predicted allele effects by quantifying S100A8 protein in inbred founder strains; and (iii) infection of C57BL/6 mice deficient for the S100a8 gene. Overall, this body of work demonstrates that systems genetics using Diversity Outbred mice can identify new (and known) QTLs and functionally relevant gene candidates that may be major regulators of complex host-pathogens interactions contributing to granuloma necrosis and acute inflammation in pulmonary TB.
Collapse
Affiliation(s)
- Daniel M. Gatti
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Anna L. Tyler
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | | | - Bulent Yener
- Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Deniz Koyuncu
- Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Metin N. Gurcan
- Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - MK Khalid Niazi
- Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - Thomas Tavolara
- Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - Adam Gower
- Clinical and Translational Science Institute, Boston University, Boston, Massachusetts, United States of America
| | - Denise Dayao
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Emily McGlone
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Melanie L. Ginese
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Aubrey Specht
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Anas Alsharaydeh
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Philipe A. Tessier
- Department of Microbiology and Immunology, Laval University School of Medicine, Quebec, Canada
| | - Sherry L. Kurtz
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Karen L. Elkins
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Igor Kramnik
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, United States of America
| | - Gillian Beamer
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| |
Collapse
|
20
|
Tang Z, Han Y, Meng Y, Li J, Qiu X, Bajinka O, Wu G, Tan Y. A bioinformatics approach to systematically analyze the molecular patterns of monkeypox virus-host cell interactions. Heliyon 2024; 10:e30483. [PMID: 38737277 PMCID: PMC11088324 DOI: 10.1016/j.heliyon.2024.e30483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 04/20/2024] [Accepted: 04/28/2024] [Indexed: 05/14/2024] Open
Abstract
Monkeypox has been spreading worldwide since May 2022, when the World Health Organization (WHO) declared the outbreak a "public health emergency of international concern." The spread of monkeypox has posed a serious threat to the health of people around the world, but few studies have been conducted, and the molecular mechanism of monkeypox after infection remains unclear. We therefore implemented a transcriptome analysis to identify signaling pathways and biomarkers in monkeypox-infected cells to help understand monkeypox-host cell interactions. In this study, datasets GSE36854 and GSE11234 were obtained from GEO. Of these, 84 significantly different genes were identified in the dataset GSE36854, followed by KEGG, GO analysis protein-protein interaction (PPI) construction, and Hub gene extraction. We also analyzed the expression regulation of hub genes and screened for drugs targeting hub genes. The results showed that monkeypox-infected cells significantly activated the cellular immune response. The top 10 hub genes are IER3, IFIT2, IL11, ZC3H12A, EREG, IER2, NFKBIE, FST, IFIT1 and AREG. AP-26113 and itraconazole can be used to counteract the inhibitory effect of monkeypox on IFIT1 and IFIT2 and serve as candidate drugs for the treatment of monkeypox virus infection. IRF1 may also be a transcription factor of IFIT. Our results provide a new entry point for understanding how monkeypox virus interacts with its host.
Collapse
Affiliation(s)
- Zhongxiang Tang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Ying Han
- Department of Stomatology, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yuting Meng
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Jiani Li
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Xiangjie Qiu
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Ousman Bajinka
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
- China-Africa Research Center for Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Guojun Wu
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
- China-Africa Research Center for Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Yurong Tan
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
- China-Africa Research Center for Infectious Diseases, School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China
| |
Collapse
|
21
|
Wang Q, Liu Y, Zhang Y, Zhang S, Zhao M, Peng Z, Xu H, Huang H. Characterization of macrophages in ischemia-reperfusion injury-induced acute kidney injury based on single-cell RNA-Seq and bulk RNA-Seq analysis. Int Immunopharmacol 2024; 130:111754. [PMID: 38428147 DOI: 10.1016/j.intimp.2024.111754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024]
Abstract
Acute kidney injury (AKI) is a complex disease, with macrophages playing a vital role in its progression. However, the mechanism of macrophage function remains unclear and strategies targeting macrophages in AKI are controversial. To address this issue, we used single-cell RNA-seq analysis to identify macrophage sub-types involved in ischemia-reperfusion-induced AKI, and then screened for associated hub genes using intersecting bulk RNA-seq data. The single-cell and bulk RNA-seq datasets were obtained from the Gene Expression Omnibus (GEO) database. Screening of differentially-expressed genes (DEGs) and pseudo-bulk DEG analyses were used to identify common hub genes. Pseudotime and trajectory analyses were performed to investigate the progression of cell differentiation. CellChat analysis was performed to reveal the crosstalk between cell clusters. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were used to identify enriched pathways in the cell clusters. Immunofluorescence and RT-PCR were preformed to validate the expression of the identified hub genes. Four hub genes, Vim, S100a6, Ier3, and Ccr1, were identified in the infiltrated macrophages between normal samples and those 3 days after ischemia-reperfusion renal injury (IRI); all were associated with the progression of IRI-induced AKI. Increased expression of Vim, S100a6, Ier3, and Ccr1 in infiltrated macrophages may be associated with inflammatory responses and may mediate crosstalk between macrophages and renal tubular epithelial cells under IRI conditions. Our results reveal that Ier3 may be critical in AKI, and that Vim, S100a6, Ier3, and Ccr1 may act as novel biomarkers and potential therapeutic targets for IRI-induced AKI.
Collapse
Affiliation(s)
- Qin Wang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuxing Liu
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Yan Zhang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| | - Siyuan Zhang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Meifang Zhao
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China.
| | - Hui Xu
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China.
| | - Hao Huang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China.
| |
Collapse
|
22
|
Tsutsumi C, Ohuchida K, Katayama N, Yamada Y, Nakamura S, Okuda S, Otsubo Y, Iwamoto C, Torata N, Horioka K, Shindo K, Mizuuchi Y, Ikenaga N, Nakata K, Nagai E, Morisaki T, Oda Y, Nakamura M. Tumor-infiltrating monocytic myeloid-derived suppressor cells contribute to the development of an immunosuppressive tumor microenvironment in gastric cancer. Gastric Cancer 2024; 27:248-262. [PMID: 38217732 DOI: 10.1007/s10120-023-01456-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/07/2023] [Indexed: 01/15/2024]
Abstract
BACKGROUND Gastric cancer (GC) is characterized by an immunosuppressive and treatment-resistant tumor immune microenvironment (TIME). Here, we investigated the roles of different immunosuppressive cell types in the development of the GC TIME. METHODS Single-cell RNA sequencing (scRNA-seq) and multiplex immunostaining of samples from untreated or immune checkpoint inhibitor (ICI)-resistant GC patients were used to examine the correlation between certain immunosuppressive cells and the prognosis of GC patients. RESULTS The results of the scRNA-seq analysis revealed that tumor-infiltrating monocytic myeloid-derived suppressor cells (TI-M-MDSCs) expressed higher levels of genes with immunosuppressive functions than other immunosuppressive cell types. Additionally, M-MDSCs in GC tissues expressed significantly higher levels of these markers than adjacent normal tissues. The M-MDSCs were most enriched in GC tissues relative to adjacent normal tissues. Among the immunosuppressive cell types assessed, the M-MDSCs were most enriched in GC tissues relative to adjacent normal tissues; moreover, their presence was most strongly associated with a poor prognosis. Immediate early response 3 (IER3), which we identified as a differentially expressed gene between M-MDSCs of GC and adjacent normal tissues, was an independent poor prognostic factor in GC patients (P = 0.0003). IER3+ M-MDSCs expressed higher levels of genes with immunosuppressive functions than IER3- M-MDSCs and were abundant in treatment-resistant GC patients. CONCLUSIONS The present study suggests that TI-M-MDSCs, especially IER3+ ones, may play a predominant role in the development of the immunosuppressive and ICI-resistant GC TIME.
Collapse
Affiliation(s)
- Chikanori Tsutsumi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Naoki Katayama
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yutaka Yamada
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shoichi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Sho Okuda
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshiki Otsubo
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Chika Iwamoto
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Nobuhiro Torata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kohei Horioka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Shindo
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yusuke Mizuuchi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Naoki Ikenaga
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Eishi Nagai
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takashi Morisaki
- Department of Cancer Immunotherapy, Fukuoka General Cancer Clinic, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
23
|
Saemann L, Wächter K, Gharpure N, Pohl S, Hoorn F, Korkmaz-Icöz S, Karck M, Veres G, Simm A, Szabó G. HTK vs. HTK-N for Coronary Endothelial Protection during Hypothermic, Oxygenated Perfusion of Hearts Donated after Circulatory Death. Int J Mol Sci 2024; 25:2262. [PMID: 38396938 PMCID: PMC10889240 DOI: 10.3390/ijms25042262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Protection of the coronary arteries during donor heart maintenance is pivotal to improve results and prevent the development of coronary allograft vasculopathy. The effect of hypothermic, oxygenated perfusion (HOP) with the traditional HTK and the novel HTK-N solution on the coronary microvasculature of donation-after-circulatory-death (DCD) hearts is known. However, the effect on the coronary macrovasculature is unknown. Thus, we maintained porcine DCD hearts by HOP with HTK or HTK-N for 4 h, followed by transplantation-equivalent reperfusion with blood for 2 h. Then, we removed the left anterior descending coronary artery (LAD) and compared the endothelial-dependent and -independent vasomotor function of both groups using bradykinin and sodium-nitroprusside (SNP). We also determined the transcriptome of LAD samples using microarrays. The endothelial-dependent relaxation was significantly better after HOP with HTK-N. The endothelial-independent relaxation was comparable between both groups. In total, 257 genes were expressed higher, and 668 genes were expressed lower in the HTK-N group. Upregulated genes/pathways were involved in endothelial and vascular smooth muscle cell preservation and heart development. Downregulated genes were related to ischemia/reperfusion injury, oxidative stress, mitochondrion organization, and immune reaction. The novel HTK-N solution preserves the endothelial function of DCD heart coronary arteries more effectively than traditional HTK.
Collapse
Affiliation(s)
- Lars Saemann
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Kristin Wächter
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
| | - Nitin Gharpure
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
| | - Sabine Pohl
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
| | - Fabio Hoorn
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Sevil Korkmaz-Icöz
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Matthias Karck
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Gábor Veres
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Andreas Simm
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
| | - Gábor Szabó
- Department of Cardiac Surgery, University Hospital Halle (Saale), University of Halle, 06120 Halle (Saale), Germany
- Department of Cardiac Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
24
|
Li N, Liang XR, Bai X, Liang XH, Dang LH, Jin QQ, Cao J, Du QX, Sun JH. Novel ratio-expressions of genes enables estimation of wound age in contused skeletal muscle. Int J Legal Med 2024; 138:197-206. [PMID: 37804331 DOI: 10.1007/s00414-023-03095-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 09/18/2023] [Indexed: 10/09/2023]
Abstract
Given that combination with multiple biomarkers may well raise the predictive value of wound age, it appears critically essential to identify new features under the limited cost. For this purpose, the present study explored whether the gene expression ratios provide unique time information as an additional indicator for wound age estimation not requiring the detection of new biomarkers and allowing full use of the available data. The expression levels of four wound-healing genes (Arid5a, Ier3, Stom, and Lcp1) were detected by real-time polymerase chain reaction, and a total of six expression ratios were calculated among these four genes. The results showed that the expression levels of four genes and six ratios of expression changed time-dependent during wound repair. The six expression ratios provided additional temporal information, distinct from the four genes analyzed separately by principal component analysis. The overall performance metrics for cross-validation and external validation of four typical prediction models were improved when six ratios of expression were added as additional input variables. Overall, expression ratios among genes provide temporal information and have excellent potential as predictive markers for wound age estimation. Combining the expression levels of genes with ratio-expression of genes may allow for more accurate estimates of the time of injury.
Collapse
Affiliation(s)
- Na Li
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, 030604, Shanxi, China
| | - Xin-Rui Liang
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, 030604, Shanxi, China
| | - Xue Bai
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, 030604, Shanxi, China
| | - Xin-Hua Liang
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, 030604, Shanxi, China
| | - Li-Hong Dang
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, 030604, Shanxi, China
| | - Qian-Qian Jin
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, 030604, Shanxi, China
| | - Jie Cao
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, 030604, Shanxi, China
| | - Qiu-Xiang Du
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, 030604, Shanxi, China.
| | - Jun-Hong Sun
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, 030604, Shanxi, China.
| |
Collapse
|
25
|
Singh P, Srivastava A, Philip L, Ahuja SK, Shivangi, Rawat C, Kutum R, Yadav J, Sood M, Chadda RK, Dash D, Vohora D, Kukreti R. Genome-wide transcriptomic and biochemical profiling of major depressive disorder: Unravelling association with susceptibility, severity, and antidepressant response. Genomics 2024; 116:110772. [PMID: 38158140 DOI: 10.1016/j.ygeno.2023.110772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/26/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
Identifying biomarkers for diagnosing Major Depressive Disorder (MDD), assessing its severity, and guiding treatment is crucial. We conducted whole genome transcriptomic study in North Indian population, and analyzed biochemical parameters. Our longitudinal study investigated gene-expression profiles from 72 drug-free MDD patients and 50 healthy controls(HCs) at baseline and 24 patients after 12-weeks of treatment. Gene expression analyses identified differentially expressed genes(DEGs) associated with MDD susceptibility, symptom severity and treatment response, independently validated by qPCR. Hierarchical clustering revealed distinct expression patterns between MDD and HCs, also between mild and severe cases. Enrichment analyses of significant DEGs revealed inflammatory, apoptosis, and immune-related pathways in MDD susceptibility, severity, and treatment response. Simultaneously, we assessed thirty biochemical parameters in the same cohort, showed significant differences between MDD and HCs in 13 parameters with monocytes, eosinophils, creatinine, SGPT, and total protein remained independent predictors of MDD in a multivariate-regression model. Our study supports the role of altered immune/inflammatory signaling in MDD pathophysiology, offering clinically relevant biochemical parameters and insights into transcriptomic gene regulation in MDD pathogenesis and treatment response.
Collapse
Affiliation(s)
- Priyanka Singh
- Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ankit Srivastava
- Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi 110007, India; Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Lini Philip
- Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi 110007, India; Department of Psychiatry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Simranpreet Kaur Ahuja
- Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi 110007, India
| | - Shivangi
- Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi 110007, India; Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Main Bawana Road, New Delhi 110042, India
| | - Chitra Rawat
- Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rintu Kutum
- Department of Computer Science, Ashoka University, Haryana 131029, India
| | - Jyoti Yadav
- Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi 110007, India
| | - Mamta Sood
- Department of Psychiatry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Rakesh Kumar Chadda
- Department of Psychiatry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Debasis Dash
- Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ritushree Kukreti
- Genomics and Molecular Medicine Unit, Council of Scientific and Industrial Research (CSIR) - Institute of Genomics and Integrative Biology (IGIB), New Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
26
|
Gatti DM, Tyler AL, Mahoney JM, Churchill GA, Yener B, Koyuncu D, Gurcan MN, Niazi M, Tavolara T, Gower AC, Dayao D, McGlone E, Ginese ML, Specht A, Alsharaydeh A, Tessier PA, Kurtz SL, Elkins K, Kramnik I, Beamer G. Systems genetics uncover new loci containing functional gene candidates in Mycobacterium tuberculosis-infected Diversity Outbred mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572738. [PMID: 38187647 PMCID: PMC10769337 DOI: 10.1101/2023.12.21.572738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Mycobacterium tuberculosis, the bacillus that causes tuberculosis (TB), infects 2 billion people across the globe, and results in 8-9 million new TB cases and 1-1.5 million deaths each year. Most patients have no known genetic basis that predisposes them to disease. We investigated the complex genetic basis of pulmonary TB by modelling human genetic diversity with the Diversity Outbred mouse population. When infected with M. tuberculosis, one-third develop early onset, rapidly progressive, necrotizing granulomas and succumb within 60 days. The remaining develop non-necrotizing granulomas and survive longer than 60 days. Genetic mapping using clinical indicators of disease, granuloma histopathological features, and immune response traits identified five new loci on mouse chromosomes 1, 2, 4, 16 and three previously identified loci on chromosomes 3 and 17. Quantitative trait loci (QTLs) on chromosomes 1, 16, and 17, associated with multiple correlated traits and had similar patterns of allele effects, suggesting these QTLs contain important genetic regulators of responses to M. tuberculosis. To narrow the list of candidate genes in QTLs, we used a machine learning strategy that integrated gene expression signatures from lungs of M. tuberculosis-infected Diversity Outbred mice with gene interaction networks, generating functional scores. The scores were then used to rank candidates for each mapped trait in each locus, resulting in 11 candidates: Ncf2, Fam20b, S100a8, S100a9, Itgb5, Fstl1, Zbtb20, Ddr1, Ier3, Vegfa, and Zfp318. Importantly, all 11 candidates have roles in infection, inflammation, cell migration, extracellular matrix remodeling, or intracellular signaling. Further, all candidates contain single nucleotide polymorphisms (SNPs), and some but not all SNPs were predicted to have deleterious consequences on protein functions. Multiple methods were used for validation including (i) a statistical method that showed Diversity Outbred mice carrying PWH/PhJ alleles on chromosome 17 QTL have shorter survival; (ii) quantification of S100A8 protein levels, confirming predicted allele effects; and (iii) infection of C57BL/6 mice deficient for the S100a8 gene. Overall, this work demonstrates that systems genetics using Diversity Outbred mice can identify new (and known) QTLs and new functionally relevant gene candidates that may be major regulators of granuloma necrosis and acute inflammation in pulmonary TB.
Collapse
Affiliation(s)
- D M Gatti
- The Jackson Laboratory, Bar Harbor, ME
| | - A L Tyler
- The Jackson Laboratory, Bar Harbor, ME
| | | | | | - B Yener
- Rensselaer Polytechnic Institute, Troy, NY
| | - D Koyuncu
- Rensselaer Polytechnic Institute, Troy, NY
| | - M N Gurcan
- Wake Forest University School of Medicine, Winston Salem, NC
| | - Mkk Niazi
- Wake Forest University School of Medicine, Winston Salem, NC
| | - T Tavolara
- Wake Forest University School of Medicine, Winston Salem, NC
| | - A C Gower
- Clinical and Translational Science Institute, Boston University, Boston, MA
| | - D Dayao
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA
| | - E McGlone
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA
| | - M L Ginese
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA
| | - A Specht
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA
| | - A Alsharaydeh
- Texas Biomedical Research Institute, San Antonio, TX
| | - P A Tessier
- Department of Microbiology and Immunology, Laval University School of Medicine, Quebec, Canada
| | - S L Kurtz
- Center for Biologics, Food and Drug Administration, Bethesda, MD
| | - K Elkins
- Center for Biologics, Food and Drug Administration, Bethesda, MD
| | - I Kramnik
- NIEDL, Boston University, Boston, MA
| | - G Beamer
- Texas Biomedical Research Institute, San Antonio, TX
| |
Collapse
|
27
|
Armstrong ND, Srinivasasainagendra V, Ammous F, Assimes TL, Beitelshees AL, Brody J, Cade BE, Ida Chen YD, Chen H, de Vries PS, Floyd JS, Franceschini N, Guo X, Hellwege JN, House JS, Hwu CM, Kardia SLR, Lange EM, Lange LA, McDonough CW, Montasser ME, O’Connell JR, Shuey MM, Sun X, Tanner RM, Wang Z, Zhao W, Carson AP, Edwards TL, Kelly TN, Kenny EE, Kooperberg C, Loos RJF, Morrison AC, Motsinger-Reif A, Psaty BM, Rao DC, Redline S, Rich SS, Rotter JI, Smith JA, Smith AV, Irvin MR, Arnett DK. Whole genome sequence analysis of apparent treatment resistant hypertension status in participants from the Trans-Omics for Precision Medicine program. Front Genet 2023; 14:1278215. [PMID: 38162683 PMCID: PMC10755672 DOI: 10.3389/fgene.2023.1278215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction: Apparent treatment-resistant hypertension (aTRH) is characterized by the use of four or more antihypertensive (AHT) classes to achieve blood pressure (BP) control. In the current study, we conducted single-variant and gene-based analyses of aTRH among individuals from 12 Trans-Omics for Precision Medicine cohorts with whole-genome sequencing data. Methods: Cases were defined as individuals treated for hypertension (HTN) taking three different AHT classes, with average systolic BP ≥ 140 or diastolic BP ≥ 90 mmHg, or four or more medications regardless of BP (n = 1,705). A normotensive control group was defined as individuals with BP < 140/90 mmHg (n = 22,079), not on AHT medication. A second control group comprised individuals who were treatment responsive on one AHT medication with BP < 140/ 90 mmHg (n = 5,424). Logistic regression with kinship adjustment using the Scalable and Accurate Implementation of Generalized mixed models (SAIGE) was performed, adjusting for age, sex, and genetic ancestry. We assessed variants using SKAT-O in rare-variant analyses. Single-variant and gene-based tests were conducted in a pooled multi-ethnicity stratum, as well as self-reported ethnic/racial strata (European and African American). Results: One variant in the known HTN locus, KCNK3, was a top finding in the multi-ethnic analysis (p = 8.23E-07) for the normotensive control group [rs12476527, odds ratio (95% confidence interval) = 0.80 (0.74-0.88)]. This variant was replicated in the Vanderbilt University Medical Center's DNA repository data. Aggregate gene-based signals included the genes AGTPBP, MYL4, PDCD4, BBS9, ERG, and IER3. Discussion: Additional work validating these loci in larger, more diverse populations, is warranted to determine whether these regions influence the pathobiology of aTRH.
Collapse
Affiliation(s)
- Nicole D. Armstrong
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | | | - Farah Ammous
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
- Survey Research Center, Institute for Social Research, Ann Arbor, MI, United States
| | - Themistocles L. Assimes
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Amber L. Beitelshees
- Division of Endocrinology, Diabetes, and Nutrition, Program for Personalized and Genomic Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jennifer Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Brian E. Cade
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Paul S. de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - James S. Floyd
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, United States
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Jacklyn N. Hellwege
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - John S. House
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Chii-Min Hwu
- Section of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Ethan M. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Caitrin W. McDonough
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - May E. Montasser
- Division of Endocrinology, Diabetes, and Nutrition, Program for Personalized and Genomic Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | | | - Megan M. Shuey
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Xiao Sun
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
| | - Rikki M. Tanner
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Zhe Wang
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
- Survey Research Center, Institute for Social Research, Ann Arbor, MI, United States
| | - April P. Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - Todd L. Edwards
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tanika N. Kelly
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL, United States
| | - Eimear E. Kenny
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Alison Motsinger-Reif
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Dabeeru C. Rao
- Division of Biostatistics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Stephen S. Rich
- Department of Public Health Sciences, Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, United States
- Survey Research Center, Institute for Social Research, Ann Arbor, MI, United States
| | - Albert V. Smith
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Donna K. Arnett
- Office of the Provost, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
28
|
Domingo-Relloso A, Joehanes R, Rodriguez-Hernandez Z, Lahousse L, Haack K, Fallin MD, Herreros-Martinez M, Umans JG, Best LG, Huan T, Liu C, Ma J, Yao C, Jerolon A, Bermudez JD, Cole SA, Rhoades DA, Levy D, Navas-Acien A, Tellez-Plaza M. Smoking, blood DNA methylation sites and lung cancer risk. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 334:122153. [PMID: 37442331 PMCID: PMC10528956 DOI: 10.1016/j.envpol.2023.122153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/07/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023]
Abstract
Altered DNA methylation (DNAm) might be a biological intermediary in the pathway from smoking to lung cancer. In this study, we investigated the contribution of differential blood DNAm to explain the association between smoking and lung cancer incidence. Blood DNAm was measured in 2321 Strong Heart Study (SHS) participants. Incident lung cancer was assessed as time to event diagnoses. We conducted mediation analysis, including validation with DNAm and paired gene expression data from the Framingham Heart Study (FHS). In the SHS, current versus never smoking and pack-years single-mediator models showed, respectively, 29 and 21 differentially methylated positions (DMPs) for lung cancer with statistically significant mediated effects (14 of 20 available, and five of 14 available, positions, replicated, respectively, in FHS). In FHS, replicated DMPs showed gene expression downregulation largely in trans, and were related to biological pathways in cancer. The multimediator model identified that DMPs annotated to the genes AHRR and IER3 jointly explained a substantial proportion of lung cancer. Thus, the association of smoking with lung cancer was partly explained by differences in baseline blood DNAm at few relevant sites. Experimental studies are needed to confirm the biological role of identified eQTMs and to evaluate potential implications for early detection and control of lung cancer.
Collapse
Affiliation(s)
- Arce Domingo-Relloso
- Integrative Epidemiology Group, Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain; Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA; Department of Statistics and Operations Research, University of Valencia, Spain.
| | - Roby Joehanes
- Population Sciences Branch, National Heart, Lung, And Blood Institute, National Institutes of Health, Bethesda, MD, USA; Framingham Heart Study, Framingham, MA, USA
| | - Zulema Rodriguez-Hernandez
- Integrative Epidemiology Group, Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
| | - Lies Lahousse
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Karin Haack
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - M Daniele Fallin
- Department of Mental Health, Johns Hopkins University, Baltimore, USA; Department of Epidemiology, Johns Hopkins University, Baltimore, USA
| | | | - Jason G Umans
- MedStar Health Research Institute, Washington DC, USA; Georgetown-Howard Universities Center for Clinical and Translational Science, Washington DC, USA
| | - Lyle G Best
- Missouri Breaks Industries and Research Inc., Eagle Butte, SD, USA
| | - Tianxiao Huan
- Framingham Heart Study, Framingham, MA, USA; University of Massachusetts Medical School, Worcester, MA, USA
| | - Chunyu Liu
- Framingham Heart Study, Framingham, MA, USA; Boston University School of Public Health, Boston, MA, USA
| | - Jiantao Ma
- Framingham Heart Study, Framingham, MA, USA; Tufts University Friedman School of Nutrition Science and Policy, Boston, MA, USA
| | - Chen Yao
- Framingham Heart Study, Framingham, MA, USA; Bristol Myers Squibb, Cambridge, MA, USA
| | - Allan Jerolon
- Université Paris Cité, CNRS, MAP5, F-75006, Paris, France
| | - Jose D Bermudez
- Department of Statistics and Operations Research, University of Valencia, Spain
| | - Shelley A Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Dorothy A Rhoades
- Stephenson Cancer Center, University of Oklahoma Health Sciences Department of Medicine, Oklahoma City, OK, USA
| | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, And Blood Institute, National Institutes of Health, Bethesda, MD, USA; Framingham Heart Study, Framingham, MA, USA
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Maria Tellez-Plaza
- Integrative Epidemiology Group, Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Madrid, Spain
| |
Collapse
|
29
|
Boulanger M, Aqrouq M, Tempé D, Kifagi C, Ristic M, Akl D, Hallal R, Carusi A, Gabellier L, de Toledo M, Sigurdsson JO, Kaoma T, Andrieu-Soler C, Forné T, Soler E, Hicheri Y, Gueret E, Vallar L, Olsen JV, Cartron G, Piechaczyk M, Bossis G. DeSUMOylation of chromatin-bound proteins limits the rapid transcriptional reprogramming induced by daunorubicin in acute myeloid leukemias. Nucleic Acids Res 2023; 51:8413-8433. [PMID: 37462077 PMCID: PMC10484680 DOI: 10.1093/nar/gkad581] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 09/09/2023] Open
Abstract
Genotoxicants have been used for decades as front-line therapies against cancer on the basis of their DNA-damaging actions. However, some of their non-DNA-damaging effects are also instrumental for killing dividing cells. We report here that the anthracycline Daunorubicin (DNR), one of the main drugs used to treat Acute Myeloid Leukemia (AML), induces rapid (3 h) and broad transcriptional changes in AML cells. The regulated genes are particularly enriched in genes controlling cell proliferation and death, as well as inflammation and immunity. These transcriptional changes are preceded by DNR-dependent deSUMOylation of chromatin proteins, in particular at active promoters and enhancers. Surprisingly, inhibition of SUMOylation with ML-792 (SUMO E1 inhibitor), dampens DNR-induced transcriptional reprogramming. Quantitative proteomics shows that the proteins deSUMOylated in response to DNR are mostly transcription factors, transcriptional co-regulators and chromatin organizers. Among them, the CCCTC-binding factor CTCF is highly enriched at SUMO-binding sites found in cis-regulatory regions. This is notably the case at the promoter of the DNR-induced NFKB2 gene. DNR leads to a reconfiguration of chromatin loops engaging CTCF- and SUMO-bound NFKB2 promoter with a distal cis-regulatory region and inhibition of SUMOylation with ML-792 prevents these changes.
Collapse
Affiliation(s)
| | - Mays Aqrouq
- IGMM, Univ. Montpellier, CNRS, Montpellier, France
| | - Denis Tempé
- IGMM, Univ. Montpellier, CNRS, Montpellier, France
| | | | - Marko Ristic
- IGMM, Univ. Montpellier, CNRS, Montpellier, France
| | - Dana Akl
- IGMM, Univ. Montpellier, CNRS, Montpellier, France
| | - Rawan Hallal
- IGMM, Univ. Montpellier, CNRS, Montpellier, France
| | - Aude Carusi
- IGMM, Univ. Montpellier, CNRS, Montpellier, France
| | - Ludovic Gabellier
- IGMM, Univ. Montpellier, CNRS, Montpellier, France
- Service d’Hématologie Clinique, CHU de Montpellier, 80 Avenue Augustin Fliche, 34091 Montpellier, France
| | | | - Jon-Otti Sigurdsson
- Proteomics Program, Novo Nordisk Foundation Center For Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Tony Kaoma
- Genomics Research Unit, Luxembourg Institute of Health, 84, Val Fleuri, L-1526 Luxembourg, Luxembourg
| | - Charlotte Andrieu-Soler
- IGMM, Univ. Montpellier, CNRS, Montpellier, France
- Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | | | - Eric Soler
- IGMM, Univ. Montpellier, CNRS, Montpellier, France
- Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | - Yosr Hicheri
- Service d’Hématologie Clinique, CHU de Montpellier, 80 Avenue Augustin Fliche, 34091 Montpellier, France
| | - Elise Gueret
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Laurent Vallar
- Genomics Research Unit, Luxembourg Institute of Health, 84, Val Fleuri, L-1526 Luxembourg, Luxembourg
| | - Jesper V Olsen
- Proteomics Program, Novo Nordisk Foundation Center For Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Guillaume Cartron
- IGMM, Univ. Montpellier, CNRS, Montpellier, France
- Service d’Hématologie Clinique, CHU de Montpellier, 80 Avenue Augustin Fliche, 34091 Montpellier, France
| | | | | |
Collapse
|
30
|
Liu Y, Chen Y, Li XH, Cao C, Zhang HX, Zhou C, Chen Y, Gong Y, Yang JX, Cheng L, Chen XD, Shen H, Xiao HM, Tan LJ, Deng HW. Dissection of Cellular Communication between Human Primary Osteoblasts and Bone Marrow Mesenchymal Stem Cells in Osteoarthritis at Single-Cell Resolution. Int J Stem Cells 2023; 16:342-355. [PMID: 37105556 PMCID: PMC10465330 DOI: 10.15283/ijsc22101] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 04/29/2023] Open
Abstract
Background and Objectives Osteoblasts are derived from bone marrow mesenchymal stem cells (BMMSCs) and play important role in bone remodeling. While our previous studies have investigated the cell subtypes and heterogeneity in osteoblasts and BMMSCs separately, cell-to-cell communications between osteoblasts and BMMSCs in vivo in humans have not been characterized. The aim of this study was to investigate the cellular communication between human primary osteoblasts and bone marrow mesenchymal stem cells. Methods and Results To investigate the cell-to-cell communications between osteoblasts and BMMSCs and identify new cell subtypes, we performed a systematic integration analysis with our single-cell RNA sequencing (scRNA-seq) transcriptomes data from BMMSCs and osteoblasts. We successfully identified a novel preosteoblasts subtype which highly expressed ATF3, CCL2, CXCL2 and IRF1. Biological functional annotations of the transcriptomes suggested that the novel preosteoblasts subtype may inhibit osteoblasts differentiation, maintain cells to a less differentiated status and recruit osteoclasts. Ligand-receptor interaction analysis showed strong interaction between mature osteoblasts and BMMSCs. Meanwhile, we found FZD1 was highly expressed in BMMSCs of osteogenic differentiation direction. WIF1 and SFRP4, which were highly expressed in mature osteoblasts were reported to inhibit osteogenic differentiation. We speculated that WIF1 and sFRP4 expressed in mature osteoblasts inhibited the binding of FZD1 to Wnt ligand in BMMSCs, thereby further inhibiting osteogenic differentiation of BMMSCs. Conclusions Our study provided a more systematic and comprehensive understanding of the heterogeneity of osteogenic cells. At the single cell level, this study provided insights into the cell-to-cell communications between BMMSCs and osteoblasts and mature osteoblasts may mediate negative feedback regulation of osteogenesis process.
Collapse
Affiliation(s)
- Ying Liu
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yan Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiao-Hua Li
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Chong Cao
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Hui-Xi Zhang
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Cui Zhou
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yu Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yun Gong
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jun-Xiao Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Liang Cheng
- Department of Orthopedics and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiang-Ding Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Hui Shen
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Hong-Mei Xiao
- School of Basic Medical Science, Central South University, Changsha, China
- Center of Reproductive Health, System Biology and Data Information, Institute of Reproductive & Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
| | - Li-Jun Tan
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Hong-Wen Deng
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
31
|
Huang J, Zhang W, Xiang R, Tan L, Liu P, Tao Z, Deng Y, Tong H, Xu Y. The early-phase transcriptome and the clinical efficacy analysis in three modes of subcutaneous immunotherapy for allergic rhinitis. World Allergy Organ J 2023; 16:100811. [PMID: 37701629 PMCID: PMC10493265 DOI: 10.1016/j.waojou.2023.100811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 07/24/2023] [Accepted: 08/12/2023] [Indexed: 09/14/2023] Open
Abstract
Background Allergen immunotherapy is the only etiological treatment for allergic rhinitis. Objective To analyze the efficacy, safety, and mechanism of subcutaneous immunotherapy (SCIT). Methods The efficacy, safety, and serum immunological changes of 3 modes of subcutaneous immunotherapy were compared. Peripheral blood mononuclear cells (PBMC) transcriptome changes were obtained on the Illumina sequencing platforms. We confirmed differentially expressed genes (DEGs) by quantitative real-time polymerase chain reaction (PCR). The DEGs were analyzed by gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and protein-protein interaction (PPI) networks. The correlation between the common DEGs and clinical indicators was analyzed by Origin 2022. Results The 3 SCITs were all effective after 1 year. The Combined Symptom and Medication Score (CSMS) and Visual Analog Score (VAS) in rush immunotherapy (RIT) are lowest after 24 and 48 weeks of treatment among the 3 groups. After treatment, the levels of sIgE, sIgE/tIgE, Th2 cytokines, Th17 cytokines, and percentage of peripheral eosinophils (EOS%) decreased significantly (P<0.05), while the levels of Th1 type cytokines did not change significantly. Transcriptome analysis identified 24, 24, and 91 DEGs at W3 and 42, 52, 175 DEGs at W7 in conventional immunotherapy (CIT), cluster immunotherapy (CLIT), and RIT groups, respectively. The pathways and functions involved in SCIT include secretion of Th1/2 cytokines, immune cell differentiation. Unlike CIT and CLIT, DEGs are also involved in T cell tolerance induction, T cell anergy, and lymphocyte anergy in RIT. CXCR1, CXCR2, and IER3 had a specific effect on reflecting the improvement of symptoms in allergic rhinitis patients with SCIT. Conclusion The clinical efficacy of RIT appeared earlier than CIT and CLIT. Clinicians can use the highly conserved gene expression profile to evaluate responses to immunotherapy.
Collapse
Affiliation(s)
- Jingyu Huang
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Zhang
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rong Xiang
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lu Tan
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Peiqiang Liu
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zezhang Tao
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, Wuhan, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuqin Deng
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huan Tong
- Wound Repair&Rehabilitation Center Department, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, China
| | - Yu Xu
- Department of Rhinology and Allergy, Otolaryngology-Head and Neck Surgery Center, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
32
|
Kuang T, Zhang L, Chai D, Chen C, Wang W. Construction of a T-cell exhaustion-related gene signature for predicting prognosis and immune response in hepatocellular carcinoma. Aging (Albany NY) 2023; 15:5751-5774. [PMID: 37354485 PMCID: PMC10333082 DOI: 10.18632/aging.204830] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/12/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a heterogeneous malignancy with a rising prevalence worldwide. Immunotherapy has been shown to improve treatment outcomes for HCC. We aimed to construct a T-cell exhaustion-related gene prognostic model (TEXPM) for HCC and to elucidate the immunologic characteristics and advantages of immunotherapy in T-cell exhaustion-Related Gene-defined HCC groups. METHODS Single-cell RNA sequencing data were used in conjunction with TCGA Differentially expressed genes (DEGs) to screen for T-cell exhaustion-Related Genes (TEXGs) for subsequent evaluation. Using univariate Cox regression analysis and LASSO regression analysis, five genes (FTL, GZMA, CD14, NPC2, and IER3) were subsequently selected for the construction of a TEXPM. Then, we evaluated the immunologic characteristics and advantages of immunotherapy in groups identified by TEXPM. RESULTS The TEXPM was formed with FTL, GZMA, CD14, NPC2, and IER3. The results of the training and validation team studies were consistent, with the low TEXPM group surviving longer than the high TEXPM group (P < 0.001). Multivariate Cox regression analysis demonstrated that TEXPM (HR: 2.347, 95%CI: 1.844-2.987; HR: 2.172, 95% CI: 1.689-2.793) was an independent prognostic variable for HCC patients. The low-TEXPM group was linked to active immunity, less aggressive phenotypes, strong infiltration of CD8+ T cells, CD4 + T cells, and M1 macrophages, and a better response to ICI treatment. A high TEXPM group, on the other hand, was associated with suppressive immunity, more aggressive phenotypes, a significant infiltration of B cells, M0 macrophages, and M2 macrophages, and a reduced response to ICI treatment. FTL is an independent prognostic variable in HCC patients and the knockdown of FTL can affect the biological behavior of hepatocellular carcinoma cells. CONCLUSIONS TEXPM is a promising prognostic biomarker connected to the immune system. Differentiating immunological and molecular features and predicting patient outcomes may be facilitated by TEXPM grouping. Furthermore, the expression of FTL was found to be an independent prognostic factor for HCC. Knockdown of FTL significantly inhibited proliferation, migration, and invasive activity in liver cancer cells.
Collapse
Affiliation(s)
- Tianrui Kuang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dongqi Chai
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
33
|
Kuperwaser F, Avital G, Vaz MJ, Noble KN, Dammann AN, Randis TM, Aronoff DM, Ratner AJ, Yanai I. Host inflammatory dynamics reveal placental immune modulation by Group B Streptococcus during pregnancy. Mol Syst Biol 2023; 19:e11021. [PMID: 36744393 PMCID: PMC9996236 DOI: 10.15252/msb.202211021] [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: 03/11/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 02/07/2023] Open
Abstract
Group B Streptococcus (GBS) is a pathobiont that can ascend to the placenta and cause adverse pregnancy outcomes, in part through production of the toxin β-hemolysin/cytolysin (β-h/c). Innate immune cells have been implicated in the response to GBS infection, but the impact of β-h/c on their response is poorly defined. We show that GBS modulates innate immune cell states by subversion of host inflammation through β-h/c, allowing worse outcomes. We used an ascending mouse model of GBS infection to measure placental cell state changes over time following infection with a β-h/c-deficient and isogenic wild type GBS strain. Transcriptomic analysis suggests that β-h/c-producing GBS elicit a worse phenotype through suppression of host inflammatory signaling in placental macrophages and neutrophils, and comparison of human placental macrophages infected with the same strains recapitulates these results. Our findings have implications for identification of new targets in GBS disease to support host defense against pathogenic challenge.
Collapse
Affiliation(s)
- Felicia Kuperwaser
- Institute for Computational MedicineNYU Grossman School of MedicineNew YorkNYUSA
| | - Gal Avital
- Institute for Computational MedicineNYU Grossman School of MedicineNew YorkNYUSA
| | - Michelle J Vaz
- Department of PediatricsNYU Grossman School of MedicineNew YorkNYUSA
| | - Kristen N Noble
- Division of Neonatology, Department of PediatricsVanderbilt University Medical CenterNashvilleTNUSA
| | - Allison N Dammann
- Renaissance School of Medicine at Stony Brook UniversityStony BrookNYUSA
| | - Tara M Randis
- Departments of Pediatrics and Molecular Medicine, Morsani School of MedicineUniversity of South FloridaFLTampaUSA
| | | | - Adam J Ratner
- Department of PediatricsNYU Grossman School of MedicineNew YorkNYUSA
- Department of MicrobiologyNYU Grossman School of MedicineNew YorkNYUSA
| | - Itai Yanai
- Institute for Computational MedicineNYU Grossman School of MedicineNew YorkNYUSA
- Department of Biochemistry and Molecular PharmacologyNYU Grossman School of MedicineNew YorkNYUSA
| |
Collapse
|
34
|
Sakharkar A, Yang J. Designing a Novel Monitoring Approach for the Effects of Space Travel on Astronauts' Health. Life (Basel) 2023; 13:life13020576. [PMID: 36836933 PMCID: PMC9964234 DOI: 10.3390/life13020576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Space exploration and extraterrestrial civilization have fascinated humankind since the earliest days of human history. It was only in the last century that humankind finally began taking significant steps towards these goals by sending astronauts into space, landing on the moon, and building the International Space Station. However, space voyage is very challenging and dangerous, and astronauts are under constant space radiation and microgravity. It has been shown that astronauts are at a high risk of developing a broad range of diseases/disorders. Thus, it is critical to develop a rapid and effective assay to monitor astronauts' health in space. In this study, gene expression and correlation patterns were analyzed for 10 astronauts (8 male and 2 female) using the publicly available microarray dataset E-GEOD-74708. We identified 218 differentially expressed genes between In-flight and Pre-flight and noticed that space travel decreased genome regulation and gene correlations across the entire genome, as well as individual signaling pathways. Furthermore, we systematically developed a shortlist of 32 genes that could be used to monitor astronauts' health during space travel. Further studies, including microgravity experiments, are warranted to optimize and validate the proposed assay.
Collapse
Affiliation(s)
- Anurag Sakharkar
- College of Arts and Science, University of Saskatchewan, 9 Campus Drive, Saskatoon, SK S7N 5A5, Canada
| | - Jian Yang
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada
- Correspondence:
| |
Collapse
|
35
|
Lopez-Tello J, Sferruzzi-Perri AN. Characterization of placental endocrine function and fetal brain development in a mouse model of small for gestational age. Front Endocrinol (Lausanne) 2023; 14:1116770. [PMID: 36843585 PMCID: PMC9950515 DOI: 10.3389/fendo.2023.1116770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
Conditions such as small for gestational age (SGA), which is defined as birthweight less than 10th percentile for gestational age can predispose to neurodevelopmental abnormalities compared to babies with normal birthweight. Fetal growth and birthweight depend on placental function, as this organ transports substrates to the developing fetus and it acts as a source of endocrine factors, including steroids and prolactins that are required for fetal development and pregnancy maintenance. To advance our knowledge on the aetiology of fetal growth disorders, the vast majority of the research has been focused on studying the transport function of the placenta, leaving practically unexplored the contribution of placental hormones in the regulation of fetal growth. Here, using mice and natural variability in fetal growth within the litter, we compared fetuses that fell on or below the 10th percentile (classified as SGA) with those that had adequate weight for their gestational age (AGA). In particular, we compared placental endocrine metabolism and hormone production, as well as fetal brain weight and expression of developmental, growth and metabolic genes between SGA and AGA fetuses. We found that compared to AGA fetuses, SGA fetuses had lower placental efficiency and reduced capacity for placental production of hormones (e.g. steroidogenic gene Cyp17a1, prolactin Prl3a1, and pregnancy-specific glycoproteins Psg21). Brain weight was reduced in SGA fetuses, although this was proportional to the reduction in overall fetal size. The expression of glucose transporter 3 (Slc2a3) was reduced despite the abundance of AKT, FOXO and ERK proteins were similar. Developmental (Sv2b and Gabrg1) and microglia genes (Ier3), as well as the pregnancy-specific glycoprotein receptor (Cd9) were lower in the brain of SGA versus AGA fetuses. In this mouse model of SGA, our results therefore demonstrate that placental endocrine dysfunction is associated with changes in fetal growth and fetal brain development.
Collapse
Affiliation(s)
- Jorge Lopez-Tello
- Centre for Trophoblast Research – Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research – Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
36
|
Novel Prediction Method Applied to Wound Age Estimation: Developing a Stacking Ensemble Model to Improve Predictive Performance Based on Multi-mRNA. Diagnostics (Basel) 2023; 13:diagnostics13030395. [PMID: 36766500 PMCID: PMC9914838 DOI: 10.3390/diagnostics13030395] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/24/2023] Open
Abstract
(1) Background: Accurate diagnosis of wound age is crucial for investigating violent cases in forensic practice. However, effective biomarkers and forecast methods are lacking. (2) Methods: Samples were collected from rats divided randomly into control and contusion groups at 0, 4, 8, 12, 16, 20, and 24 h post-injury. The characteristics of concern were nine mRNA expression levels. Internal validation data were used to train different machine learning algorithms, namely random forest (RF), support vector machine (SVM), multilayer perceptron (MLP), gradient boosting (GB), and stochastic gradient descent (SGD), to predict wound age. These models were considered the base learners, which were then applied to developing 26 stacking ensemble models combining two, three, four, or five base learners. The best-performing stacking model and base learner were evaluated through external validation data. (3) Results: The best results were obtained using a stacking model of RF + SVM + MLP (accuracy = 92.85%, area under the receiver operating characteristic curve (AUROC) = 0.93, root-mean-square-error (RMSE) = 1.06 h). The wound age prediction performance of the stacking models was also confirmed for another independent dataset. (4) Conclusions: We illustrate that machine learning techniques, especially ensemble algorithms, have a high potential to be used to predict wound age. According to the results, the strategy can be applied to other types of forensic forecasts.
Collapse
|
37
|
Ismailova A, Salehi-Tabar R, Dimitrov V, Memari B, Barbier C, White JH. Identification of a forkhead box protein transcriptional network induced in human neutrophils in response to inflammatory stimuli. Front Immunol 2023; 14:1123344. [PMID: 36756115 PMCID: PMC9900176 DOI: 10.3389/fimmu.2023.1123344] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 01/25/2023] Open
Abstract
Introduction Neutrophils represent the largest proportion of circulating leukocytes and, in response to inflammatory stimuli, are rapidly recruited to sites of infection where they neutralize pathogens. Methods and results We have identified a novel neutrophil transcription network induced in response to inflammatory stimuli. We performed the first RNAseq analysis of human neutrophils exposed to lipopolysaccharide (LPS), followed by a meta-analysis of our dataset and previously published studies of LPS-challenged neutrophils. This revealed a robustly enhanced transcriptional network driven by forkhead box (FOX) transcription factors. The network is enriched in genes encoding proinflammatory cytokines and transcription factors, including MAFF and ATF3, which are implicated in responses to stress, survival and inflammation. Expression of transcription factors FOXP1 and FOXP4 is induced in neutrophils exposed to inflammatory stimuli, and potential FOXP1/FOXP4 binding sites were identified in several genes in the network, all located in chromatin regions consistent with neutrophil enhancer function. Chromatin immunoprecipitation (ChIP) assays in neutrophils confirmed enhanced binding of FOXP4, but not FOXP1, to multiple sites in response to LPS. Binding to numerous motifs and transactivation of network genes were also observed when FOXP proteins were transiently expressed in HEK293 cells. In addition to LPS, the transcriptional network is induced by other inflammatory stimuli, indicating it represents a general neutrophil response to inflammation. Discussion Collectively, these findings reveal a role for the FOXP4 transcription network as a regulator of responses to inflammatory stimuli in neutrophils.
Collapse
Affiliation(s)
- Aiten Ismailova
- Department of Physiology, McGill University, Montreal, QC, Canada
| | | | - Vassil Dimitrov
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Babak Memari
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Camille Barbier
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - John H. White
- Department of Physiology, McGill University, Montreal, QC, Canada,Department of Medicine, McGill University, Montreal, QC, Canada,*Correspondence: John H. White,
| |
Collapse
|
38
|
Lyubetskaya A, Rabe B, Fisher A, Lewin A, Neuhaus I, Brett C, Brett T, Pereira E, Golhar R, Kebede S, Font-Tello A, Mosure K, Van Wittenberghe N, Mavrakis KJ, MacIsaac K, Chen BJ, Drokhlyansky E. Assessment of spatial transcriptomics for oncology discovery. CELL REPORTS METHODS 2022; 2:100340. [PMID: 36452860 PMCID: PMC9701619 DOI: 10.1016/j.crmeth.2022.100340] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 08/05/2022] [Accepted: 10/21/2022] [Indexed: 06/17/2023]
Abstract
Tumor heterogeneity is a major challenge for oncology drug discovery and development. Understanding of the spatial tumor landscape is key to identifying new targets and impactful model systems. Here, we test the utility of spatial transcriptomics (ST) for oncology discovery by profiling 40 tissue sections and 80,024 capture spots across a diverse set of tissue types, sample formats, and RNA capture chemistries. We verify the accuracy and fidelity of ST by leveraging matched pathology analysis, which provides a ground truth for tissue section composition. We then use spatial data to demonstrate the capture of key tumor depth features, identifying hypoxia, necrosis, vasculature, and extracellular matrix variation. We also leverage spatial context to identify relative cell-type locations showing the anti-correlation of tumor and immune cells in syngeneic cancer models. Lastly, we demonstrate target identification approaches in clinical pancreatic adenocarcinoma samples, highlighting tumor intrinsic biomarkers and paracrine signaling.
Collapse
Affiliation(s)
- Anna Lyubetskaya
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Brian Rabe
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Andrew Fisher
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Anne Lewin
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Isaac Neuhaus
- Research and Early Development, Bristol Myers Squibb Company, Route 206 & Province Line Road, Princeton, NJ 08543, USA
| | - Constance Brett
- Aggregate Genius, Inc., 560 Fulford-Ganges Road, Salt Spring Island, BC V8K 2K1, Canada
| | - Todd Brett
- Aggregate Genius, Inc., 560 Fulford-Ganges Road, Salt Spring Island, BC V8K 2K1, Canada
| | - Ethel Pereira
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Ryan Golhar
- Research and Early Development, Bristol Myers Squibb Company, Route 206 & Province Line Road, Princeton, NJ 08543, USA
| | - Sami Kebede
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Alba Font-Tello
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Kathy Mosure
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Nicholas Van Wittenberghe
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Konstantinos J. Mavrakis
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Kenzie MacIsaac
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Benjamin J. Chen
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| | - Eugene Drokhlyansky
- Research and Early Development, Bristol Myers Squibb Company, 100 Binney Street, Cambridge, MA 02142, USA
| |
Collapse
|
39
|
Fox HS, Niu M, Morsey BM, Lamberty BG, Emanuel K, Periyasamy P, Callen S, Acharya A, Kubik G, Eudy J, Guda C, Dyavar SR, Fletcher CV, Byrareddy SN, Buch S. Morphine suppresses peripheral responses and transforms brain myeloid gene expression to favor neuropathogenesis in SIV infection. Front Immunol 2022; 13:1012884. [PMID: 36466814 PMCID: PMC9709286 DOI: 10.3389/fimmu.2022.1012884] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/31/2022] [Indexed: 11/18/2022] Open
Abstract
The twin pandemics of opioid abuse and HIV infection can have devastating effects on physiological systems, including on the brain. Our previous work found that morphine increased the viral reservoir in the brains of treated SIV-infected macaques. In this study, we investigated the interaction of morphine and SIV to identify novel host-specific targets using a multimodal approach. We probed systemic parameters and performed single-cell examination of the targets for infection in the brain, microglia and macrophages. Morphine treatment created an immunosuppressive environment, blunting initial responses to infection, which persisted during antiretroviral treatment. Antiretroviral drug concentrations and penetration into the cerebrospinal fluid and brain were unchanged by morphine treatment. Interestingly, the transcriptional signature of both microglia and brain macrophages was transformed to one of a neurodegenerative phenotype. Notably, the expression of osteopontin, a pleiotropic cytokine, was significantly elevated in microglia. This was especially notable in the white matter, which is also dually affected by HIV and opioids. Increased osteopontin expression was linked to numerous HIV neuropathogenic mechanisms, including those that can maintain a viral reservoir. The opioid morphine is detrimental to SIV/HIV infection, especially in the brain.
Collapse
Affiliation(s)
- Howard S. Fox
- Departments of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States,*Correspondence: Howard S. Fox,
| | - Meng Niu
- Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Brenda M. Morsey
- Departments of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Benjamin G. Lamberty
- Departments of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Katy Emanuel
- Departments of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Palsamy Periyasamy
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shannon Callen
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Arpan Acharya
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Gregory Kubik
- The Genomics Core Facility, University of Nebraska Medical Center, Omaha, NE, United States
| | - James Eudy
- Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Chittibabu Guda
- Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shetty Ravi Dyavar
- The Antiviral Pharmacology Laboratory, University of Nebraska Medical Center, Omaha, NE, United States
| | - Courtney V. Fletcher
- The Antiviral Pharmacology Laboratory, University of Nebraska Medical Center, Omaha, NE, United States
| | - Siddappa N. Byrareddy
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shilpa Buch
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
40
|
Zhai F, Wang J, Yang W, Ye M, Jin X. The E3 Ligases in Cervical Cancer and Endometrial Cancer. Cancers (Basel) 2022; 14:5354. [PMID: 36358773 PMCID: PMC9658772 DOI: 10.3390/cancers14215354] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 07/28/2023] Open
Abstract
Endometrial (EC) and cervical (CC) cancers are the most prevalent malignancies of the female reproductive system. There is a global trend towards increasing incidence and mortality, with a decreasing age trend. E3 ligases label substrates with ubiquitin to regulate their activity and stability and are involved in various cellular functions. Studies have confirmed abnormal expression or mutations of E3 ligases in EC and CC, indicating their vital roles in the occurrence and progression of EC and CC. This paper provides an overview of the E3 ligases implicated in EC and CC and discusses their underlying mechanism. In addition, this review provides research advances in the target of ubiquitination processes in EC and CC.
Collapse
Affiliation(s)
- Fengguang Zhai
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo 315211, China
| | - Jie Wang
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo 315211, China
| | - Weili Yang
- Department of Gynecology, The Affiliated People’s Hospital of Ningbo University, Ningbo 315040, China
| | - Meng Ye
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo 315211, China
| | - Xiaofeng Jin
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Medical School of Ningbo University, Ningbo 315211, China
| |
Collapse
|
41
|
Ross BX, Jia L, Kong D, Wang T, Yao J, Hager HM, Abcouwer SF, Zacks DN. Hypoxia-Inducible Factor-1α in Rods Is Neuroprotective Following Retinal Detachment. Invest Ophthalmol Vis Sci 2022; 63:7. [PMID: 36223101 PMCID: PMC9583748 DOI: 10.1167/iovs.63.11.7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/22/2022] [Indexed: 02/02/2023] Open
Abstract
Purpose Following retinal detachment (RD) photoreceptors (PRs) sustain hypoxic stress and eventually die. Hypoxia-inducible factor-1α (HIF-1α) plays a central role in cellular adaptation to hypoxia. The purpose of this study is to determine the necessity of HIF-1α on PR cell survival after RD. Methods Experimental RD was created in mice by injection of hyaluronic acid (1%) into the subretinal space. Mice with conditional HIF-1α knockout in rods (denoted as HIF-1αΔrod) were used. HIF-1α expression in retinas was measured real-time polymerase chain reaction (RT-PCR) and Western blotting. PR cell death after RD was evaluated using TUNEL assay. Optical coherence tomography (OCT) and histology were used to evaluate retinal layer thicknesses and PR cell densities. A hypoxia signaling pathway PCR array was used to examine the expression of HIF-1α target genes after RD. Results HIF-1α protein levels were significantly increased after RD, and depletion of HIF-1α in rods blunted this increase. A compensatory increase of HIF-2α protein was observed in HIF-1αΔrod mice. Conditional knockout (cKO) of HIF-1α in rods did not lead to any morphologic change in attached retinas but resulted in significantly increased PR cell loss after RD. HIF-1α cKO in rods altered the responses to retinal detachment for 25 out of 83 HIF-1α target genes that were highly enriched for genes involved in glycolysis. Conclusions Rod-derived HIF-1α plays a key role in the PR response to RD, mediating the transcriptional activity of a battery of genes to promote PR cell survival.
Collapse
Affiliation(s)
- Bing X. Ross
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Lin Jia
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Dejuan Kong
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Tiantian Wang
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Jingyu Yao
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Heather M. Hager
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Steven F. Abcouwer
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - David N. Zacks
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| |
Collapse
|
42
|
Vasileva NS, Kuligina EV, Dymova MA, Savinovskaya YI, Zinchenko ND, Ageenko AB, Mishinov SV, Dome AS, Stepanov GA, Richter VA, Semenov DV. Transcriptome Changes in Glioma Cells Cultivated under Conditions of Neurosphere Formation. Cells 2022; 11:cells11193106. [PMID: 36231068 PMCID: PMC9563256 DOI: 10.3390/cells11193106] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Glioma is the most common and heterogeneous primary brain tumor. The development of a new relevant preclinical models is necessary. As research moves from cultures of adherent gliomas to a more relevant model, neurospheres, it is necessary to understand the changes that cells undergo at the transcriptome level. In the present work, we used three patient-derived gliomas and two immortalized glioblastomas, while their cultivation was carried out under adherent culture and neurosphere (NS) conditions. When comparing the transcriptomes of monolayer (ML) and NS cell cultures, we used Enrichr genes sets enrichment analysis to describe transcription factors (TFs) and the pathways involved in the formation of glioma NS. It was observed that NS formation is accompanied by the activation of five common gliomas of TFs, SOX2, UBTF, NFE2L2, TCF3 and STAT3. The sets of transcripts controlled by TFs MYC and MAX were suppressed in NS. Upregulated genes are involved in the processes of the epithelial-mesenchymal transition, cancer stemness, invasion and migration of glioma cells. However, MYC/MAX-dependent downregulated genes are involved in translation, focal adhesion and apical junction. Furthermore, we found three EGFR and FGFR signaling feedback regulators common to all analyzed gliomas-SPRY4, ERRFI1, and RAB31-which can be used for creating new therapeutic strategies of suppressing the invasion and progression of gliomas.
Collapse
Affiliation(s)
- Natalia S. Vasileva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue 8, Novosibirsk 630090, Russia
| | - Elena V. Kuligina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue 8, Novosibirsk 630090, Russia
| | - Maya A. Dymova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue 8, Novosibirsk 630090, Russia
| | - Yulya I. Savinovskaya
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue 8, Novosibirsk 630090, Russia
| | - Nikita D. Zinchenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue 8, Novosibirsk 630090, Russia
| | - Alisa B. Ageenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue 8, Novosibirsk 630090, Russia
| | - Sergey V. Mishinov
- Novosibirsk Research Institute of Traumatology and Orthopedics n.a. Ya.L. Tsivyan, Department of Neurosurgery, Frunze Street 17, Novosibirsk 630091, Russia
| | - Anton S. Dome
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue 8, Novosibirsk 630090, Russia
| | - Grigory A. Stepanov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue 8, Novosibirsk 630090, Russia
| | - Vladimir A. Richter
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue 8, Novosibirsk 630090, Russia
| | - Dmitry V. Semenov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue 8, Novosibirsk 630090, Russia
- Correspondence: ; Tel.: +73-833635189
| |
Collapse
|
43
|
Aczél T, Benczik B, Ágg B, Körtési T, Urbán P, Bauer W, Gyenesei A, Tuka B, Tajti J, Ferdinandy P, Vécsei L, Bölcskei K, Kun J, Helyes Z. Disease- and headache-specific microRNA signatures and their predicted mRNA targets in peripheral blood mononuclear cells in migraineurs: role of inflammatory signalling and oxidative stress. J Headache Pain 2022; 23:113. [PMID: 36050647 PMCID: PMC9438144 DOI: 10.1186/s10194-022-01478-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Migraine is a primary headache with genetic susceptibility, but the pathophysiological mechanisms are poorly understood, and it remains an unmet medical need. Earlier we demonstrated significant differences in the transcriptome of migraineurs' PBMCs (peripheral blood mononuclear cells), suggesting the role of neuroinflammation and mitochondrial dysfunctions. Post-transcriptional gene expression is regulated by miRNA (microRNA), a group of short non-coding RNAs that are emerging biomarkers, drug targets, or drugs. MiRNAs are emerging biomarkers and therapeutics; however, little is known about the miRNA transcriptome in migraine, and a systematic comparative analysis has not been performed so far in migraine patients. METHODS We determined miRNA expression of migraineurs' PBMC during (ictal) and between (interictal) headaches compared to age- and sex-matched healthy volunteers. Small RNA sequencing was performed from the PBMC, and mRNA targets of miRNAs were predicted using a network theoretical approach by miRNAtarget.com™. Predicted miRNA targets were investigated by Gene Ontology enrichment analysis and validated by comparing network metrics to differentially expressed mRNA data. RESULTS In the interictal PBMC samples 31 miRNAs were differentially expressed (DE) in comparison to healthy controls, including hsa-miR-5189-3p, hsa-miR-96-5p, hsa-miR-3613-5p, hsa-miR-99a-3p, hsa-miR-542-3p. During headache attacks, the top DE miRNAs as compared to the self-control samples in the interictal phase were hsa-miR-3202, hsa-miR-7855-5p, hsa-miR-6770-3p, hsa-miR-1538, and hsa-miR-409-5p. MiRNA-mRNA target prediction and pathway analysis indicated several mRNAs related to immune and inflammatory responses (toll-like receptor and cytokine receptor signalling), neuroinflammation and oxidative stress, also confirmed by mRNA transcriptomics. CONCLUSIONS We provide here the first evidence for disease- and headache-specific miRNA signatures in the PBMC of migraineurs, which might help to identify novel targets for both prophylaxis and attack therapy.
Collapse
Affiliation(s)
- Timea Aczél
- Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, Molecular Pharmacology Research Group, Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - Bettina Benczik
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Bence Ágg
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Tamás Körtési
- MTA-SZTE Neuroscience Research Group, University of Szeged, Szeged, Hungary
- Faculty of Health Sciences and Social Studies, University of Szeged, Szeged, Hungary
| | - Péter Urbán
- Szentágothai Research Centre, Bioinformatics Research Group, Genomics and Bioinformatics Core Facility, University of Pécs, Pécs, Hungary
| | - Witold Bauer
- Szentágothai Research Centre, Bioinformatics Research Group, Genomics and Bioinformatics Core Facility, University of Pécs, Pécs, Hungary
| | - Attila Gyenesei
- Szentágothai Research Centre, Bioinformatics Research Group, Genomics and Bioinformatics Core Facility, University of Pécs, Pécs, Hungary
| | - Bernadett Tuka
- MTA-SZTE Neuroscience Research Group, University of Szeged, Szeged, Hungary
- Faculty of Health Sciences and Social Studies, University of Szeged, Szeged, Hungary
| | - János Tajti
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Péter Ferdinandy
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - László Vécsei
- MTA-SZTE Neuroscience Research Group, University of Szeged, Szeged, Hungary
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Kata Bölcskei
- Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, Molecular Pharmacology Research Group, Centre for Neuroscience, University of Pécs, Pécs, Hungary
| | - József Kun
- Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, Molecular Pharmacology Research Group, Centre for Neuroscience, University of Pécs, Pécs, Hungary
- Szentágothai Research Centre, Bioinformatics Research Group, Genomics and Bioinformatics Core Facility, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School & Szentágothai Research Centre, Molecular Pharmacology Research Group, Centre for Neuroscience, University of Pécs, Pécs, Hungary.
- PharmInVivo Ltd., Pécs, Hungary.
- Department of Pharmacology and Pharmacotherapy, University of Pécs Medical School, Szigeti út 12, 7624, Pécs, Hungary.
| |
Collapse
|
44
|
Campos JHC, Alves GV, Maricato JT, Braconi CT, Antoneli FM, Janini LMR, Briones MRS. The epitranscriptome of Vero cells infected with SARS-CoV-2 assessed by direct RNA sequencing reveals m6A pattern changes and DRACH motif biases in viral and cellular RNAs. Front Cell Infect Microbiol 2022; 12:906578. [PMID: 36051243 PMCID: PMC9425070 DOI: 10.3389/fcimb.2022.906578] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
The epitranscriptomics of the SARS-CoV-2 infected cell reveals its response to viral replication. Among various types of RNA nucleotide modifications, the m6A is the most common and is involved in several crucial processes of RNA intracellular location, maturation, half-life and translatability. This epitranscriptome contains a mixture of viral RNAs and cellular transcripts. In a previous study we presented the analysis of the SARS-CoV-2 RNA m6A methylation based on direct RNA sequencing and characterized DRACH motif mutations in different viral lineages. Here we present the analysis of the m6A transcript methylation of Vero cells (derived from African Green Monkeys) and Calu-3 cells (human) upon infection by SARS-CoV-2 using direct RNA sequencing data. Analysis of these data by nonparametric statistics and two computational methods (m6anet and EpiNano) show that m6A levels are higher in RNAs of infected cells. Functional enrichment analysis reveals increased m6A methylation of transcripts involved in translation, peptide and amine metabolism. This analysis allowed the identification of differentially methylated transcripts and m6A unique sites in the infected cell transcripts. Results here presented indicate that the cell response to viral infection not only changes the levels of mRNAs, as previously shown, but also its epitranscriptional pattern. Also, transcriptome-wide analysis shows strong nucleotide biases in DRACH motifs of cellular transcripts, both in Vero and Calu-3 cells, which use the signature GGACU whereas in viral RNAs the signature is GAACU. We hypothesize that the differences of DRACH motif biases, might force the convergent evolution of the viral genome resulting in better adaptation to target sequence preferences of writer, reader and eraser enzymes. To our knowledge, this is the first report on m6A epitranscriptome of the SARS-CoV-2 infected Vero cells by direct RNA sequencing, which is the sensu stricto RNA-seq.
Collapse
Affiliation(s)
- João H. C. Campos
- Center for Medical Bioinformatics, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Gustavo V. Alves
- Center for Medical Bioinformatics, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Juliana T. Maricato
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Carla T. Braconi
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Fernando M. Antoneli
- Center for Medical Bioinformatics, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Luiz Mario R. Janini
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Marcelo R. S. Briones
- Center for Medical Bioinformatics, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| |
Collapse
|
45
|
Zhu L, Zhu C, Wang J, Yang R, Zhao X. The association between DNA methylation of 6p21.33 and AHRR in blood and coronary heart disease in Chinese population. BMC Cardiovasc Disord 2022; 22:370. [PMID: 35964014 PMCID: PMC9375073 DOI: 10.1186/s12872-022-02766-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 07/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Early detection could significantly improve the prognosis of coronary heart disease (CHD). In-invitro diagnostic technique may provide a solution when sufficient biomarkers could be identified. Pertinent associations between blood-based aberrant DNA methylation and smoking, the pathogenesis of atherosclerosis, and CHD have been robustly demonstrated and replicated, but that studies in Chinese populations are rare. The blood-based methylation of aryl-hydrocarbon receptor repressor (AHRR) cg05575921 and 6p21.33 cg06126421 has been associated with cardiovascular mortality in Caucasians. Here, we aim to investigate whether the AHRR and 6p21.33 methylation in the blood is associated with CHD in the Chinese population. METHODS In this case-control study, 180 CHD patients recruited at their first registration in our study center, and 184 controls randomly selected from the people who participated in the annual health examination were enrolled. Methylation intensities of 19 CpG sites, including AHRR cg05575921, 6p21.33 cg06126421, and their flanking CpG sites, were quantified by mass spectrometry. The association between methylation intensities and CHD was estimated by logistic regression analyses adjusted for covariant. RESULTS Compared to the controls, lower methylation of 6p21.33_CpG_4.5/cg06126421 was independently associated with increased odds of being a CHD patient (OR per - 10% methylation = 1.42 after adjustment for age, gender, and batch effect; p = 0.032 by multiple testing corrections). No association between blood-based AHRR methylation and CHD was found. CONCLUSIONS 6p21.33 methylation exhibits a significant association with CHD. The combination of 6p21.33 methylation and conventional risk factors might be an intermediate step towards the early detection of CHD.
Collapse
Affiliation(s)
- Liya Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chao Zhu
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, West District, Beijing, 100050, China
| | - Jinxin Wang
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Rongxi Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Xiaojing Zhao
- Military Translational Medicine Lab, Medical Innovation Research Division, Chinese PLA General Hospital, Beijing, 100853, China. .,Beijing Key Laboratory of Chronic Heart Failure Precision Medicine, Medical Innovation Research Division, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
46
|
Pacheco NL, Noren Hooten N, Zhang Y, Prince CS, Mode NA, Ezike N, Becker KG, Zonderman AB, Evans MK. Sex-specific transcriptome differences in a middle-aged frailty cohort. BMC Geriatr 2022; 22:651. [PMID: 35945487 PMCID: PMC9361278 DOI: 10.1186/s12877-022-03326-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Frailty is a clinical syndrome described as reduced physiological reserve and increased vulnerability. Typically examined in older adults, recent work shows frailty occurs in middle-aged individuals and is associated with increased mortality. Previous investigation of global transcriptome changes in a middle-aged cohort from the Healthy Aging in Neighborhoods of Diversity across the Life Span (HANDLS) study demonstrated inflammatory genes and pathways were significantly altered by frailty status and race. Transcriptome differences in frailty by sex remain unclear. We sought to discover novel genes and pathways associated with sex and frailty in a diverse middle-aged cohort using RNA-Sequencing. METHODS Differential gene expression and pathway analyses were performed in peripheral blood mononuclear cells for 1) frail females (FRAF, n = 4) vs non-frail females (NORF, n = 4), 2) frail males (FRAM, n = 4) vs non-frail males (NORM, n = 4), 3) FRAM vs FRAF, and 4) NORM vs NORF. We evaluated exclusive significant genes and pathways, as well as overlaps, between the comparison groups. RESULTS Over 80% of the significant genes exclusive to FRAF vs NORF, FRAM vs NORM, and FRAM vs FRAF, respectively, were novel and associated with various biological functions. Pathways exclusive to FRAF vs NORF were associated with reduced inflammation, while FRAM vs NORM exclusive pathways were related to aberrant musculoskeletal physiology. Pathways exclusive to FRAM vs FRAF were associated with reduced cell cycle regulation and activated catabolism and Coronavirus pathogenesis. CONCLUSIONS Our results indicate sex-specific transcriptional changes occur in middle-aged frailty, enhancing knowledge on frailty progression and potential therapeutic targets to prevent frailty.
Collapse
Affiliation(s)
- Natasha L Pacheco
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yongqing Zhang
- Laboratory of Genetics and Genomics, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Calais S Prince
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nicolle A Mode
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ngozi Ezike
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Kevin G Becker
- Laboratory of Genetics and Genomics, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
47
|
Xiao L, Cheng H, Cai H, Wei Y, Zan G, Feng X, Liu C, Li L, Huang L, Wang F, Chen X, Zou Y, Yang X. Associations of Heavy Metals with Activities of Daily Living Disability: An Epigenome-Wide View of DNA Methylation and Mediation Analysis. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:87009. [PMID: 36036794 PMCID: PMC9423034 DOI: 10.1289/ehp10602] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 07/07/2022] [Accepted: 08/15/2022] [Indexed: 06/12/2023]
Abstract
BACKGROUND Exposure to heavy metals has been reported to be associated with multiple diseases. However, direct associations and potential mechanisms of heavy metals with physical disability remain unclear. OBJECTIVES We aimed to quantify associations of heavy metals with physical disability and further explore the potential mechanisms of DNA methylation on the genome scale. METHODS A cross-sectional study of 4,391 older adults was conducted and activities of daily living (ADL) disability were identified using a 14-item scale questionnaire including basic and instrumental activities to assess the presence of disability (yes or no) rated on a scale of dependence. Odds ratios (ORs) and 95% confidence intervals (CI) were estimated to quantify associations between heavy metals and ADL disability prevalence using multivariate logistic regression and Bayesian kernel machine regression (BKMR) models. Whole blood-derived DNA methylation was measured using the HumanMethylationEPIC BeadChip array. An ADL disability-related epigenome-wide DNA methylation association study (EWAS) was performed among 212 sex-matched ADL disability cases and controls, and mediation analysis was further applied to explore potential mediators of DNA methylation. RESULTS Each 1-standard deviation (SD) higher difference in log10-transformed manganese, copper, arsenic, and cadmium level was significantly associated with a 14% (95% CI: 1.05, 1.24), 16% (95% CI:1.07, 1.26), 22% (95% CI:1.13, 1.33), and 15% (95% CI:1.06, 1.26) higher odds of ADL disability, which remained significant in the multiple-metal and BKMR models. A total of 85 differential DNA methylation sites were identified to be associated with ADL disability prevalence, among which methylation level at cg220000984 and cg23012519 (annotated to IRGM and PKP3) mediated 31.0% and 31.2% of manganese-associated ADL disability prevalence, cg06723863 (annotated to ESRP2) mediated 32.4% of copper-associated ADL disability prevalence, cg24433124 (nearest to IER3) mediated 15.8% of arsenic-associated ADL disability prevalence, and cg07905190 and cg17485717 (annotated to FREM1 and TCP11L1) mediated 21.5% and 30.5% of cadmium-associated ADL disability prevalence (all p<0.05). DISCUSSION Our findings suggested that heavy metals contributed to higher prevalence of ADL disability and that locus-specific DNA methylation are partial mediators, providing potential biomarkers for further cellular mechanism studies. https://doi.org/10.1289/EHP10602.
Collapse
Affiliation(s)
- Lili Xiao
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Hong Cheng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Haiqing Cai
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yue Wei
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Gaohui Zan
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiuming Feng
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Chaoqun Liu
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Longman Li
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Lulu Huang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Fei Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xing Chen
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Yunfeng Zou
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaobo Yang
- Department of Occupational Health and Environmental Health, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
48
|
Construction and validation of a novel apoptosis-associated prognostic signature related to osteosarcoma metastasis and immune infiltration. Transl Oncol 2022; 22:101452. [PMID: 35598382 PMCID: PMC9126984 DOI: 10.1016/j.tranon.2022.101452] [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: 03/16/2022] [Revised: 04/20/2022] [Accepted: 05/08/2022] [Indexed: 11/22/2022] Open
Abstract
Osteosarcoma is one of the most aggressive diseases which often develops metastasis. Apoptosis relates to the recurrence and metastasis of osteosarcoma and the related signature could predict the prognosis of patients. A novel apoptosis-associated prognosis signature related to osteosarcoma metastasis and immune infiltration has been developed. The signature could help to predict the prognosis of osteosarcoma patients and serve as the potential targets for anti-cancer treatment.
Background Apoptosis played vital roles in the formation and progression of osteosarcoma. However, no studies elucidated the prognostic relationships between apoptosis-associated genes (AAGs) and osteosarcoma. Methods The differentially expressed genes associated with osteosarcoma metastasis and apoptosis were identified from GEO and MSigDB databases. The apoptosis-associated prognostic signature was established through univariate and multivariate cox regression analyses. The Kaplan–Meier (KM) survival curve, ROC curve and nomogram were constructed to investigate the predictive value of this signature. CIBERSORT algorithm and ssGSEA were used to explore the relationships between immune infiltration and AAG signature. The above results were validated in another GEO dataset and the expression of AAGs was also validated in osteosarcoma patient samples by immunohistochemistry. Results HSPB1 and IER3 were involved in AAG signature. In training and validation datasets, apoptosis-associated risk scores were negatively related to patient survival rates and the AAG signature was regarded as the independent prognostic factor. ROC and calibration curves demonstrated the signature and nomogram were reliable. GSEA revealed the signature related to immune-associated pathways. ssGSEA indicated that one immune cell and three immune functions were significantly dysregulated. The immunohistochemistry analyses of patients’ samples revealed that AAGs were significantly differently expressed between metastasis and non-metastasis osteosarcomas. Conclusions The present study identified and validated a novel apoptosis-associated prognostic signature related to osteosarcoma metastasis. It could serve as the potential biomarker and therapeutic targets for osteosarcoma in the future.
Collapse
|
49
|
Wu Y, Huang Y, Zhou C, Wang H, Wang Z, Wu J, Nie S, Deng X, Sun J, Gao X. A Novel Necroptosis-Related Prognostic Signature of Glioblastoma Based on Transcriptomics Analysis and Single Cell Sequencing Analysis. Brain Sci 2022; 12:brainsci12080988. [PMID: 35892430 PMCID: PMC9460316 DOI: 10.3390/brainsci12080988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Glioblastoma (GBM) is the most common and deadly brain tumor. The clinical significance of necroptosis (NCPS) genes in GBM is unclear. The goal of this study is to reveal the potential prognostic NCPS genes associated with GBM, elucidate their functions, and establish an effective prognostic model for GBM patients. Methods: Firstly, the NCPS genes in GBM were identified by single-cell analysis of the GSE182109 dataset in the GEO database and weighted co-expression network analysis (WGCNA) of The Cancer Genome Atlas (TCGA) data. Three machine learning algorithms (Lasso, SVM-RFE, Boruta) combined with COX regression were used to build prognostic models. The subsequent analysis included survival, immune microenvironments, and mutations. Finally, the clinical significance of NCPS in GBM was explored by constructing nomograms. Results: We constructed a GBM prognostic model composed of NCPS-related genes, including CTSD, AP1S1, YWHAG, and IER3, which were validated to have good performance. According to the above prognostic model, GBM patients in the TCGA and CGGA groups could be divided into two groups according to NCPS, with significant differences in survival analysis between the two groups and a markedly worse prognostic status in the high NCPS group (p < 0.001). In addition, the high NCPS group had higher levels of immune checkpoint-related gene expression, suggesting that they may be more likely to benefit from immunotherapy. Conclusions: Four genes (CTSD, AP1S1, YWHAG, and IER3) were screened through three machine learning algorithms to construct a prognostic model for GBM. These key and novel diagnostic markers may become new targets for diagnosing and treating patients with GBM.
Collapse
Affiliation(s)
- Yiwen Wu
- Department of Neurosurgery, Ningbo First Hospital, Zhejiang University School of Medicine, Ningbo 315010, China; (Y.W.); (C.Z.); (H.W.); (Z.W.); (J.W.); (S.N.); (X.D.); (J.S.)
| | - Yi Huang
- Department of Neurosurgery, Ningbo First Hospital, Zhejiang University School of Medicine, Ningbo 315010, China; (Y.W.); (C.Z.); (H.W.); (Z.W.); (J.W.); (S.N.); (X.D.); (J.S.)
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315010, China
- Medical Research Center, Ningbo First Hospital, Ningbo 315010, China
- Correspondence: (Y.H.); (X.G.)
| | - Chenhui Zhou
- Department of Neurosurgery, Ningbo First Hospital, Zhejiang University School of Medicine, Ningbo 315010, China; (Y.W.); (C.Z.); (H.W.); (Z.W.); (J.W.); (S.N.); (X.D.); (J.S.)
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315010, China
- Medical Research Center, Ningbo First Hospital, Ningbo 315010, China
| | - Haifeng Wang
- Department of Neurosurgery, Ningbo First Hospital, Zhejiang University School of Medicine, Ningbo 315010, China; (Y.W.); (C.Z.); (H.W.); (Z.W.); (J.W.); (S.N.); (X.D.); (J.S.)
| | - Zhepei Wang
- Department of Neurosurgery, Ningbo First Hospital, Zhejiang University School of Medicine, Ningbo 315010, China; (Y.W.); (C.Z.); (H.W.); (Z.W.); (J.W.); (S.N.); (X.D.); (J.S.)
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315010, China
| | - Jiawei Wu
- Department of Neurosurgery, Ningbo First Hospital, Zhejiang University School of Medicine, Ningbo 315010, China; (Y.W.); (C.Z.); (H.W.); (Z.W.); (J.W.); (S.N.); (X.D.); (J.S.)
| | - Sheng Nie
- Department of Neurosurgery, Ningbo First Hospital, Zhejiang University School of Medicine, Ningbo 315010, China; (Y.W.); (C.Z.); (H.W.); (Z.W.); (J.W.); (S.N.); (X.D.); (J.S.)
| | - Xinpeng Deng
- Department of Neurosurgery, Ningbo First Hospital, Zhejiang University School of Medicine, Ningbo 315010, China; (Y.W.); (C.Z.); (H.W.); (Z.W.); (J.W.); (S.N.); (X.D.); (J.S.)
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo 315010, China
| | - Jie Sun
- Department of Neurosurgery, Ningbo First Hospital, Zhejiang University School of Medicine, Ningbo 315010, China; (Y.W.); (C.Z.); (H.W.); (Z.W.); (J.W.); (S.N.); (X.D.); (J.S.)
| | - Xiang Gao
- Department of Neurosurgery, Ningbo First Hospital, Zhejiang University School of Medicine, Ningbo 315010, China; (Y.W.); (C.Z.); (H.W.); (Z.W.); (J.W.); (S.N.); (X.D.); (J.S.)
- Correspondence: (Y.H.); (X.G.)
| |
Collapse
|
50
|
Nguyen TM, Le HL, Hwang KB, Hong YC, Kim JH. Predicting High Blood Pressure Using DNA Methylome-Based Machine Learning Models. Biomedicines 2022; 10:biomedicines10061406. [PMID: 35740428 PMCID: PMC9220060 DOI: 10.3390/biomedicines10061406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 12/12/2022] Open
Abstract
DNA methylation modification plays a vital role in the pathophysiology of high blood pressure (BP). Herein, we applied three machine learning (ML) algorithms including deep learning (DL), support vector machine, and random forest for detecting high BP using DNA methylome data. Peripheral blood samples of 50 elderly individuals were collected three times at three visits for DNA methylome profiling. Participants who had a history of hypertension and/or current high BP measure were considered to have high BP. The whole dataset was randomly divided to conduct a nested five-group cross-validation for prediction performance. Data in each outer training set were independently normalized using a min–max scaler, reduced dimensionality using principal component analysis, then fed into three predictive algorithms. Of the three ML algorithms, DL achieved the best performance (AUPRC = 0.65, AUROC = 0.73, accuracy = 0.69, and F1-score = 0.73). To confirm the reliability of using DNA methylome as a biomarker for high BP, we constructed mixed-effects models and found that 61,694 methylation sites located in 15,523 intragenic regions and 16,754 intergenic regions were significantly associated with BP measures. Our proposed models pioneered the methodology of applying ML and DNA methylome data for early detection of high BP in clinical practices.
Collapse
Affiliation(s)
- Thi Mai Nguyen
- Department of Integrative Bioscience & Biotechnology, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Korea;
| | - Hoang Long Le
- Department of Computer Science & Engineering, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Korea;
| | - Kyu-Baek Hwang
- School of Computer Science & Engineering, Soongsil University, 369 Sangdo-ro, Dongjak-gu, Seoul 06978, Korea;
| | - Yun-Chul Hong
- Department of Preventive Medicine, College of Medicine, Seoul National University, Seoul 03080, Korea;
- Institute of Environmental Medicine, Seoul National University Medical Research Center, Seoul 03080, Korea
| | - Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Korea;
- Correspondence: ; Tel.: +82-2-3408-3655
| |
Collapse
|