1
|
Zimmermann S, Roomp K, Meyer H, Mathew A, Struck MF, Blüher M, Martin HNG, Keller M, Landgraf K, Körner A, Hoffmann A, Böttcher Y, Biemann K, Ghosh A, Wolfrum C, Noé F, Isermann B, Schneider JG, Biemann R. Association of Lifestyle-Induced Weight Loss With Gene Expression in Subcutaneous Adipose Tissue in Metabolic Syndrome. J Diabetes 2025; 17:e70083. [PMID: 40229590 PMCID: PMC11996622 DOI: 10.1111/1753-0407.70083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
AIMS Lifestyle-induced weight loss (LIWL) is considered an effective therapy for the treatment of metabolic syndrome (MetS). The role of differentially expressed genes (DEGs) in adipose tissue function and in the success of LIWL in MetS is still unclear. We investigated the effect of 6 months of LIWL on transcriptional regulation in subcutaneous adipose tissue (SAT). Aiming to identify a LIWL-associated "gene signature" in SAT, DEGs were fitted into a linear regression model. MATERIALS AND METHODS The study is embedded in a prospective, two-arm, controlled, monocentric, randomized, 6-month interventional trial in individuals with MetS following LIWL. The trial included 43 nonsmoking, nondiabetic men aged 45-55 years with MetS. RESULTS In total, we identified 642 DEGs in SAT after 6 months of LIWL. The identified DEGs were validated in two cross-sectional cohorts analyzing SAT from individuals with and without obesity. Gene enrichment analysis of the DEGs revealed the strongest association with cholesterol metabolic processes. Accordingly, DEGs were correlated with the lipid parameters HDL cholesterol, LDL cholesterol, and triglycerides in corresponding serum samples. We identified 3 genes with an AUC of 0.963 (95% CI: 0.906-1.0) associated with a loss of more than 10% of initial body weight that was maintained for at least 12 months after LIWL, namely SUMO3 (Small ubiquitin-related modifier 3), PRKG2 (Protein Kinase CGMP-Dependent 2), and ADAP2 (ArfGAP with Dual PH Domains 2). CONCLUSION In summary, we have identified DEGs in SAT after LIWL, which may play an important role in metabolic functions. In particular, altered gene expression in SAT may predict sustained weight loss.
Collapse
Affiliation(s)
- Silke Zimmermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular DiagnosticsUniversity of LeipzigLeipzigGermany
| | - Kirsten Roomp
- Luxembourg Centre for Systems Biomedicine (LCSB)University of LuxembourgLuxembourgLuxembourg
| | - Hans‐Jonas Meyer
- Diagnostic and Interventional RadiologyUniversity of Leipzig Faculty of MedicineLeipzigGermany
| | - Akash Mathew
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular DiagnosticsUniversity of LeipzigLeipzigGermany
| | - Manuel Florian Struck
- Department of Anesthesiology and Intensive Care MedicineUniversity Hospital LeipzigLeipzigGermany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
- Medical Department III—Endocrinology, Nephrology, and RheumatologyLeipzig University Medical CenterLeipzigGermany
- German Center for Child and Adolescent Health (DZKJ)Leipzig/Dresden Partner SiteLeipzigGermany
| | - Hugo N. G. Martin
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Maria Keller
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
- Medical Department III—Endocrinology, Nephrology, and RheumatologyLeipzig University Medical CenterLeipzigGermany
| | - Kathrin Landgraf
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & AdolescentsUniversity of LeipzigLeipzigGermany
| | - Antje Körner
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
- German Center for Child and Adolescent Health (DZKJ)Leipzig/Dresden Partner SiteLeipzigGermany
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & AdolescentsUniversity of LeipzigLeipzigGermany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI‐MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital LeipzigLeipzigGermany
| | - Yvonne Böttcher
- University of OsloInstitute of Clinical Medicine, Department of Clinical Molecular Biology, EpiGenOsloNorway
- Medical Division, EpiGenAkershus University HospitalLørenskogNorway
| | - Kathleen Biemann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular DiagnosticsUniversity of LeipzigLeipzigGermany
| | - Adhideb Ghosh
- Institute of Food, Nutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Falko Noé
- Institute of Food, Nutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular DiagnosticsUniversity of LeipzigLeipzigGermany
| | - Jochen G. Schneider
- Luxembourg Centre for Systems Biomedicine (LCSB)University of LuxembourgLuxembourgLuxembourg
- Department of Internal Medicine IISaarland University Medical Center at Homburg/SaarHomburgGermany
- Centre Hospitalier Emile MayrischEsch sur AlzetteLuxembourg
| | - Ronald Biemann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular DiagnosticsUniversity of LeipzigLeipzigGermany
| |
Collapse
|
2
|
Irani Shemirani M. Transcriptional markers classifying Escherichia coli and Staphylococcus aureus induced sepsis in adults: A data-driven approach. PLoS One 2024; 19:e0305920. [PMID: 38968271 PMCID: PMC11226107 DOI: 10.1371/journal.pone.0305920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 06/06/2024] [Indexed: 07/07/2024] Open
Abstract
Sepsis is a life-threatening condition mainly caused by gram-negative and gram-positive bacteria. Understanding the type of causative agent in the early stages is essential for precise antibiotic therapy. This study sought to identify a host gene set capable of distinguishing between sepsis induced by gram-negative bacteria; Escherichia coli and gram-positive bacteria; Staphylococcus aureus in community-onset adult patients. In the present study, microarray expression information was used to apply the Least Absolute Shrinkage and Selection Operator (Lasso) technique to select the predictive gene set for classifying sepsis induced by E. coli or S. aureus pathogens. We identified 25 predictive genes, including LILRA5 and TNFAIP6, which had previously been associated with sepsis in other research. Using these genes, we trained a logistic regression classifier to distinguish whether a sample contains an E. coli or S. aureus infection or belongs to a healthy control group, and subsequently assessed its performance. The classifier achieved an Area Under the Curve (AUC) of 0.96 for E. coli and 0.98 for S. aureus-induced sepsis, and perfect discrimination (AUC of 1) for healthy controls from the other conditions in a 10-fold cross-validation. The genes demonstrated an AUC of 0.75 in distinguishing between sepsis patients with E. coli and S. aureus pathogens. These findings were further confirmed in two distinct independent validation datasets which gave high prediction AUC ranging from 0.72-0.87 and 0.62 in distinguishing three groups of participants and two groups of patients respectively. These genes were significantly enriched in the immune system, cytokine signaling in immune system, innate immune system, and interferon signaling. Transcriptional patterns in blood can differentiate patients with E. coli-induced sepsis from those with S. aureus-induced sepsis. These diagnostic markers, upon validation in larger trials, may serve as a foundation for a reliable differential diagnostics assay.
Collapse
Affiliation(s)
- Mahnaz Irani Shemirani
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
3
|
Zhang S, Zhang M, Zhang L, Wang Z, Tang S, Yang X, Li Z, Feng J, Qin X. Identification of Y‒linked biomarkers and exploration of immune infiltration of normal-appearing gray matter in multiple sclerosis by bioinformatic analysis. Heliyon 2024; 10:e28085. [PMID: 38515685 PMCID: PMC10956066 DOI: 10.1016/j.heliyon.2024.e28085] [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: 05/15/2023] [Revised: 03/03/2024] [Accepted: 03/12/2024] [Indexed: 03/23/2024] Open
Abstract
Background The knowledge of normal‒appearing cortical gray matter (NAGM) in multiple sclerosis (MS) remains unclear. In this study, we aimed to identify diagnostic biomarkers and explore the immune infiltration characteristics of NAGM in MS through bioinformatic analysis and validation in vivo. Methods Differentially expressed genes (DEGs) were analyzed. Subsequently, the functional pathways of the DEGs were determined. After screening the overlapping DEGs of MS with two machine learning methods, the biomarkers' efficacy and the expression levels of overlapping DEGs were calculated. Quantitative reverse transcription polymerase chain reaction (qRT‒PCR) identified the robust diagnostic biomarkers. Additionally, infiltrating immune cell populations were estimated and correlated with the biomarkers. Finally, the characteristics of immune infiltration of NAGM from MS were evaluated. Results A total of 98 DEGs were identified. They participated in sensory transduction of the olfactory system, synaptic signaling, and immune responses. Nine overlapping genes were screened by machine learning methods. After verified by ROC curve, four genes, namely HLA‒DRB1, RPS4Y1, EIF1AY and USP9Y, were screened as candidate biomarkers. The mRNA expression of RPS4Y1 and USP9Y was significantly lower in MS patients than that in the controls. They were selected as the robust diagnostic biomarkers for male MS patients. RPS4Y1 and USP9Y were both positively correlated with memory B cells. Moreover, naive CD4+ T cells and monocytes were increased in the NAGM of MS patients compared with those in controls. Conclusions Low expressed Y‒linked genes, RPS4Y1 and USP9Y, were identified as diagnostic biomarkers for MS in male patients. The inhomogeneity of immune cells in NAGM might exacerbate intricate interplay between the CNS and the immune system in the MS.
Collapse
Affiliation(s)
| | | | - Lei Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Zijie Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Shi Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Xiaolin Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Zhizhong Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Jinzhou Feng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, 1st Youyi Road, Yuzhong District, Chongqing, 400016, China
| |
Collapse
|
4
|
Burel JG, Wang W, Wuhrer M, Dedicoat M, Fletcher TE, Cunningham AF, O'Shea MK. IgG glycosylation associates with risk of progression from latent to active tuberculosis. J Infect 2024; 88:106115. [PMID: 38309308 DOI: 10.1016/j.jinf.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/23/2024] [Accepted: 01/28/2024] [Indexed: 02/05/2024]
Abstract
OBJECTIVES Glycosylation motifs shape antibody structure, stability and antigen affinity and play an important role in antibody localization and function. Serum IgG glycosylation profiles are significantly altered in infectious diseases, including tuberculosis (TB), but have not been studied in the context of progression from latent to active TB. METHODS We performed a longitudinal study of paired bulk IgG glycosylation and transcriptomic profiling in blood from individuals with active TB (ATB) or latent TB infection (LTBI) before and after treatment. RESULTS We identified that a combination of two IgG1 glycosylation traits were sufficient to distinguish ATB from LTBI with high specificity and sensitivity, prior to, and after treatment. Importantly, these two features positively correlated with previously defined cellular and RNA signatures of ATB risk in LTBI, namely monocyte to lymphocyte ratio and the expression of interferon (IFN)-associated gene signature of progression (IFN-risk signature) in blood prior to treatment. Additional glycosylation features at higher prevalence in LTBI individuals with high expression of the IFN-risk signature prior to treatment included fucosylation on IgG1, IgG2 and IgG3. CONCLUSIONS Together, our results demonstrate that bulk IgG glycosylation features could be useful in stratifying the risk of LTBI reactivation and progression to ATB.
Collapse
Affiliation(s)
- Julie G Burel
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Wenjun Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Martin Dedicoat
- Department of Infection, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Thomas E Fletcher
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; Academic Department of Military Medicine, Royal Centre for Defence Medicine, Birmingham, UK
| | - Adam F Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Matthew K O'Shea
- Department of Infection, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Academic Department of Military Medicine, Royal Centre for Defence Medicine, Birmingham, UK; Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| |
Collapse
|
5
|
Wang Y, Ma X, Xu E, Huang Z, Yang C, Zhu K, Dong Y, Zhang C. Identifying squalene epoxidase as a metabolic vulnerability in high-risk osteosarcoma using an artificial intelligence-derived prognostic index. Clin Transl Med 2024; 14:e1586. [PMID: 38372422 PMCID: PMC10875711 DOI: 10.1002/ctm2.1586] [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: 11/10/2023] [Revised: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Osteosarcoma (OSA) presents a clinical challenge and has a low 5-year survival rate. Currently, the lack of advanced stratification models makes personalized therapy difficult. This study aims to identify novel biomarkers to stratify high-risk OSA patients and guide treatment. METHODS We combined 10 machine-learning algorithms into 101 combinations, from which the optimal model was established for predicting overall survival based on transcriptomic profiles for 254 samples. Alterations in transcriptomic, genomic and epigenomic landscapes were assessed to elucidate mechanisms driving poor prognosis. Single-cell RNA sequencing (scRNA-seq) unveiled genes overexpressed in OSA cells as potential therapeutic targets, one of which was validated via tissue staining, knockdown and pharmacological inhibition. We characterized changes in multiple phenotypes, including proliferation, colony formation, migration, invasion, apoptosis, chemosensitivity and in vivo tumourigenicity. RNA-seq and Western blotting elucidated the impact of squalene epoxidase (SQLE) suppression on signalling pathways. RESULTS The artificial intelligence-derived prognostic index (AIDPI), generated by our model, was an independent prognostic biomarker, outperforming clinicopathological factors and previously published signatures. Incorporating the AIDPI with clinical factors into a nomogram improved predictive accuracy. For user convenience, both the model and nomogram are accessible online. Patients in the high-AIDPI group exhibited chemoresistance, coupled with overexpression of MYC and SQLE, increased mTORC1 signalling, disrupted PI3K-Akt signalling, and diminished immune infiltration. ScRNA-seq revealed high expression of MYC and SQLE in OSA cells. Elevated SQLE expression correlated with chemoresistance and worse outcomes in OSA patients. Therapeutically, silencing SQLE suppressed OSA malignancy and enhanced chemosensitivity, mediated by cholesterol depletion and suppression of the FAK/PI3K/Akt/mTOR pathway. Furthermore, the SQLE-specific inhibitor FR194738 demonstrated anti-OSA effects in vivo and exhibited synergistic effects with chemotherapeutic agents. CONCLUSIONS AIDPI is a robust biomarker for identifying the high-risk subset of OSA patients. The SQLE protein emerges as a metabolic vulnerability in these patients, providing a target with translational potential.
Collapse
Affiliation(s)
- Yongjie Wang
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
- Proteomics and Cancer Cell Signaling Group, German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Xiaolong Ma
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
| | - Enjie Xu
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
| | - Zhen Huang
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
| | - Chen Yang
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Kunpeng Zhu
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
| | - Yang Dong
- Department of OrthopaedicsShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai Jiao Tong UniversityShanghaiP. R. China
| | - Chunlin Zhang
- Department of Orthopaedic SurgeryShanghai Tenth People's Hospital, School of Medicine, Tongji UniversityShanghaiP. R. China
- Institute of Bone Tumor Affiliated to Tongji University School of MedicineShanghaiP. R. China
| |
Collapse
|
6
|
Lee J, Sternberg H, Bignone PA, Murai J, Malik NN, West MD, Larocca D. Clonal and Scalable Endothelial Progenitor Cell Lines from Human Pluripotent Stem Cells. Biomedicines 2023; 11:2777. [PMID: 37893151 PMCID: PMC10604251 DOI: 10.3390/biomedicines11102777] [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: 09/07/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) can be used as a renewable source of endothelial cells for treating cardiovascular disease and other ischemic conditions. Here, we present the derivation and characterization of a panel of distinct clonal embryonic endothelial progenitor cells (eEPCs) lines that were differentiated from human embryonic stem cells (hESCs). The hESC line, ESI-017, was first partially differentiated to produce candidate cultures from which eEPCs were cloned. Endothelial cell identity was assessed by transcriptomic analysis, cell surface marker expression, immunocytochemical marker analysis, and functional analysis of cells and exosomes using vascular network forming assays. The transcriptome of the eEPC lines was compared to various adult endothelial lines as well as various non-endothelial cells including both adult and embryonic origins. This resulted in a variety of distinct cell lines with functional properties of endothelial cells and strong transcriptomic similarity to adult endothelial primary cell lines. The eEPC lines, however, were distinguished from adult endothelium by their novel pattern of embryonic gene expression. We demonstrated eEPC line scalability of up to 80 population doublings (pd) and stable long-term expansion of over 50 pd with stable angiogenic properties at late passage. Taken together, these data support the finding that hESC-derived clonal eEPC lines are a potential source of scalable therapeutic cells and cell products for treating cardiovascular disease. These eEPC lines offer a highly promising resource for the development of further preclinical studies aimed at therapeutic interventions.
Collapse
Affiliation(s)
- Jieun Lee
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Alameda, CA 94501, USA; (H.S.); (P.A.B.); (N.N.M.); (D.L.)
| | - Hal Sternberg
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Alameda, CA 94501, USA; (H.S.); (P.A.B.); (N.N.M.); (D.L.)
| | - Paola A. Bignone
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Alameda, CA 94501, USA; (H.S.); (P.A.B.); (N.N.M.); (D.L.)
| | - James Murai
- Advanced Cell Technology, Alameda, CA 94502, USA
| | - Nafees N. Malik
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Alameda, CA 94501, USA; (H.S.); (P.A.B.); (N.N.M.); (D.L.)
| | | | - Dana Larocca
- AgeX Therapeutics, Inc., 1101 Marina Village Parkway, Alameda, CA 94501, USA; (H.S.); (P.A.B.); (N.N.M.); (D.L.)
| |
Collapse
|
7
|
Xavier A, Maltby VE, Ewing E, Campagna MP, Burnard SM, Tegner JN, Slee M, Butzkueven H, Kockum I, Kular L, Ausimmune/AusLong Investigators Group, Jokubaitis VG, Kilpatrick T, Alfredsson L, Jagodic M, Ponsonby AL, Taylor BV, Scott RJ, Lea RA, Lechner-Scott J. DNA Methylation Signatures of Multiple Sclerosis Occur Independently of Known Genetic Risk and Are Primarily Attributed to B Cells and Monocytes. Int J Mol Sci 2023; 24:12576. [PMID: 37628757 PMCID: PMC10454485 DOI: 10.3390/ijms241612576] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/20/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Epigenetic mechanisms can regulate how DNA is expressed independently of sequence and are known to be associated with various diseases. Among those epigenetic mechanisms, DNA methylation (DNAm) is influenced by genotype and the environment, making it an important molecular interface for studying disease etiology and progression. In this study, we examined the whole blood DNA methylation profiles of a large group of people with (pw) multiple sclerosis (MS) compared to those of controls. We reveal that methylation differences in pwMS occur independently of known genetic risk loci and show that they more strongly differentiate disease (AUC = 0.85, 95% CI 0.82-0.89, p = 1.22 × 10-29) than known genetic risk loci (AUC = 0.72, 95% CI: 0.66-0.76, p = 9.07 × 10-17). We also show that methylation differences in MS occur predominantly in B cells and monocytes and indicate the involvement of cell-specific biological pathways. Overall, this study comprehensively characterizes the immune cell-specific epigenetic architecture of MS.
Collapse
Affiliation(s)
- Alexandre Xavier
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (A.X.); (S.M.B.); (R.J.S.)
| | - Vicki E. Maltby
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (V.E.M.); (R.A.L.)
- Department of Neurology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| | - Ewoud Ewing
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Maria Pia Campagna
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (M.P.C.); (H.B.); (V.G.J.)
| | - Sean M. Burnard
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (A.X.); (S.M.B.); (R.J.S.)
| | - Jesper N. Tegner
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia;
- Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
- Unit of Computational Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, L8:05, 17176 Stockholm, Sweden
- Science for Life Laboratory, Tomtebodavagen 23A, 17165 Solna, Sweden
| | - Mark Slee
- College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia;
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (M.P.C.); (H.B.); (V.G.J.)
- MSBase Foundation, Melbourne, VIC 3004, Australia
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Lara Kular
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | | | - Vilija G. Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; (M.P.C.); (H.B.); (V.G.J.)
| | - Trevor Kilpatrick
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3052, Australia; (T.K.); (A.-L.P.)
| | - Lars Alfredsson
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden; (E.E.); (I.K.); (L.K.); (L.A.); (M.J.)
| | - Anne-Louise Ponsonby
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC 3052, Australia; (T.K.); (A.-L.P.)
- National Centre for Epidemiology and Public Health, Australian National University, Canberra, ACT 2601, Australia
| | - Bruce V. Taylor
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia;
| | - Rodney J. Scott
- School of Biomedical Sciences and Pharmacy, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (A.X.); (S.M.B.); (R.J.S.)
- Department of Molecular Genetics, Pathology North, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| | - Rodney A. Lea
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (V.E.M.); (R.A.L.)
- Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Kelvin Grove, QLD 4059, Australia
| | - Jeannette Lechner-Scott
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, NSW 2305, Australia; (V.E.M.); (R.A.L.)
- Department of Neurology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
8
|
Kang H, Hasselbeck S, Taškova K, Wang N, Oosten LNV, Mrowka R, Utikal J, Andrade-Navarro MA, Wang J, Wölfl S, Cheng X. Development of a next-generation endogenous OCT4 inducer and its anti-aging effect in vivo. Eur J Med Chem 2023; 257:115513. [PMID: 37253308 DOI: 10.1016/j.ejmech.2023.115513] [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: 03/24/2023] [Revised: 05/05/2023] [Accepted: 05/23/2023] [Indexed: 06/01/2023]
Abstract
The identification of small molecules capable of replacing transcription factors has been a longstanding challenge in the generation of human chemically induced pluripotent stem cells (iPSCs). Recent studies have shown that ectopic expression of OCT4, one of the master pluripotency regulators, compromised the developmental potential of resulting iPSCs, This highlights the importance of finding endogenous OCT4 inducers for the generation of clinical-grade human iPSCs. Through a cell-based high throughput screen, we have discovered several new OCT4-inducing compounds (O4Is). In this work, we prepared metabolically stable analogues, including O4I4, which activate endogenous OCT4 and associated signaling pathways in various cell lines. By combining these with a transcription factor cocktail consisting of SOX2, KLF4, MYC, and LIN28 (referred to as "CSKML") we achieved to reprogram human fibroblasts into a stable and authentic pluripotent state without the need for exogenous OCT4. In Caenorhabditis elegans and Drosophila, O4I4 extends lifespan, suggesting the potential application of OCT4-inducing compounds in regenerative medicine and rejuvenation therapy.
Collapse
Affiliation(s)
- Han Kang
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Germany
| | - Sebastian Hasselbeck
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt am Main, Germany
| | - Katerina Taškova
- Faculty of Biology, Johannes Gutenberg University Mainz, Germany
| | - Nessa Wang
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Germany
| | - Luuk N van Oosten
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Germany
| | - Ralf Mrowka
- Experimentelle Nephrologie, KIM III, Universitätsklinikum, Jena, Germany
| | - Jochen Utikal
- Skin Cancer Unit (G300), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Jichang Wang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Stefan Wölfl
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Germany
| | - Xinlai Cheng
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Germany; Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt am Main, Germany; Frankfurt Cancer Institute, Germany.
| |
Collapse
|
9
|
Landscape of Well-Coordinated Fracture Healing in a Mouse Model Using Molecular and Cellular Analysis. Int J Mol Sci 2023; 24:ijms24043569. [PMID: 36834981 PMCID: PMC9964763 DOI: 10.3390/ijms24043569] [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: 12/27/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
The success of fracture healing relies on overlapping but coordinated cellular and molecular events. Characterizing an outline of differential gene regulation throughout successful healing is essential for identifying crucial phase-specific markers and may serve as the basis for engineering these in challenging healing situations. This study analyzed the healing progression of a standard closed femoral fracture model in C57BL/6N (age = 8 weeks) wild-type male mice. The fracture callus was assessed across various days post fracture (D = days 0, 3, 7, 10, 14, 21, and 28) by microarray, with D0 serving as a control. Histological analyses were carried out on samples from D7 until D28 to support the molecular findings. Microarray analysis revealed a differential regulation of immune response, angiogenesis, ossification, extracellular matrix regulation, mitochondrial and ribosomal genes during healing. In-depth analysis showed differential regulation of mitochondrial and ribosomal genes during the initial phase of healing. Furthermore, the differential gene expression showed an essential role of Serpin Family F Member 1 over the well-known Vascular Endothelial Growth Factor in angiogenesis, especially during the inflammatory phase. The significant upregulation of matrix metalloproteinase 13 and bone sialoprotein from D3 until D21 asserts their importance in bone mineralization. The study also shows type I collagen around osteocytes located in the ossified region at the periosteal surface during the first week of healing. Histological analysis of matrix extracellular phosphoglycoprotein and extracellular signal-regulated kinase stressed their roles in bone homeostasis and the physiological bone-healing process. This study reveals previously unknown and novel candidates, that could serve as a target for specific time points in healing and to remedy cases of impaired healing.
Collapse
|
10
|
Systems level analysis of sex-dependent gene expression changes in Parkinson's disease. NPJ Parkinsons Dis 2023; 9:8. [PMID: 36681675 PMCID: PMC9867746 DOI: 10.1038/s41531-023-00446-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
Parkinson's disease (PD) is a heterogeneous disorder, and among the factors which influence the symptom profile, biological sex has been reported to play a significant role. While males have a higher age-adjusted disease incidence and are more frequently affected by muscle rigidity, females present more often with disabling tremors. The molecular mechanisms involved in these differences are still largely unknown, and an improved understanding of the relevant factors may open new avenues for pharmacological disease modification. To help address this challenge, we conducted a meta-analysis of disease-associated molecular sex differences in brain transcriptomics data from case/control studies. Both sex-specific (alteration in only one sex) and sex-dimorphic changes (changes in both sexes, but with opposite direction) were identified. Using further systems level pathway and network analyses, coordinated sex-related alterations were studied. These analyses revealed significant disease-associated sex differences in mitochondrial pathways and highlight specific regulatory factors whose activity changes can explain downstream network alterations, propagated through gene regulatory cascades. Single-cell expression data analyses confirmed the main pathway-level changes observed in bulk transcriptomics data. Overall, our analyses revealed significant sex disparities in PD-associated transcriptomic changes, resulting in coordinated modulations of molecular processes. Among the regulatory factors involved, NR4A2 has already been reported to harbor rare mutations in familial PD and its pharmacological activation confers neuroprotective effects in toxin-induced models of Parkinsonism. Our observations suggest that NR4A2 may warrant further research as a potential adjuvant therapeutic target to address a subset of pathological molecular features of PD that display sex-associated profiles.
Collapse
|
11
|
Hephzibah Cathryn R, Udhaya Kumar S, Younes S, Zayed H, George Priya Doss C. A review of bioinformatics tools and web servers in different microarray platforms used in cancer research. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 131:85-164. [PMID: 35871897 DOI: 10.1016/bs.apcsb.2022.05.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Over the past decade, conventional lab work strategies have gradually shifted from being limited to a laboratory setting towards a bioinformatics era to help manage and process the vast amounts of data generated by omics technologies. The present work outlines the latest contributions of bioinformatics in analyzing microarray data and their application to cancer. We dissect different microarray platforms and their use in gene expression in cancer models. We highlight how computational advances empowered the microarray technology in gene expression analysis. The study on protein-protein interaction databases classified into primary, derived, meta-database, and prediction databases describes the strategies to curate and predict novel interaction networks in silico. In addition, we summarize the areas of bioinformatics where neural graph networks are currently being used, such as protein functions, protein interaction prediction, and in silico drug discovery and development. We also discuss the role of deep learning as a potential tool in the prognosis, diagnosis, and treatment of cancer. Integrating these resources efficiently, practically, and ethically is likely to be the most challenging task for the healthcare industry over the next decade; however, we believe that it is achievable in the long term.
Collapse
Affiliation(s)
- R Hephzibah Cathryn
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - S Udhaya Kumar
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Salma Younes
- Department of Biomedical Sciences, College of Health and Sciences, Qatar University, QU Health, Doha, Qatar
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health and Sciences, Qatar University, QU Health, Doha, Qatar
| | - C George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India.
| |
Collapse
|
12
|
Graf M, Interlandi M, Moreno N, Holdhof D, Göbel C, Melcher V, Mertins J, Albert TK, Kastrati D, Alfert A, Holsten T, de Faria F, Meisterernst M, Rossig C, Warmuth-Metz M, Nowak J, Meyer Zu Hörste G, Mayère C, Nef S, Johann P, Frühwald MC, Dugas M, Schüller U, Kerl K. Single-cell transcriptomics identifies potential cells of origin of MYC rhabdoid tumors. Nat Commun 2022; 13:1544. [PMID: 35318328 PMCID: PMC8941154 DOI: 10.1038/s41467-022-29152-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/26/2022] [Indexed: 11/30/2022] Open
Abstract
Rhabdoid tumors (RT) are rare and highly aggressive pediatric neoplasms. Their epigenetically-driven intertumoral heterogeneity is well described; however, the cellular origin of RT remains an enigma. Here, we establish and characterize different genetically engineered mouse models driven under the control of distinct promoters and being active in early progenitor cell types with diverse embryonic onsets. From all models only Sox2-positive progenitor cells give rise to murine RT. Using single-cell analyses, we identify distinct cells of origin for the SHH and MYC subgroups of RT, rooting in early stages of embryogenesis. Intra- and extracranial MYC tumors harbor common genetic programs and potentially originate from fetal primordial germ cells (PGCs). Using PGC specific Smarcb1 knockout mouse models we validate that MYC RT originate from these progenitor cells. We uncover an epigenetic imbalance in MYC tumors compared to PGCs being sustained by epigenetically-driven subpopulations. Importantly, treatments with the DNA demethylating agent decitabine successfully impair tumor growth in vitro and in vivo. In summary, our work sheds light on the origin of RT and supports the clinical relevance of DNA methyltransferase inhibitors against this disease. Rhabdoid tumors (RT) are aggressive paediatric cancers with yet unknown cells of origin. Here, the authors establish genetically engineered mouse models of RT and, using single-cell RNA-seq and epigenomics, identify potential cells of origin for the SHH and MYC subtypes.
Collapse
Affiliation(s)
- Monika Graf
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Marta Interlandi
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany.,Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Natalia Moreno
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Dörthe Holdhof
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.,Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Carolin Göbel
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.,Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Viktoria Melcher
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Julius Mertins
- Department of Neurology, Schlosspark-Klinik, 14059, Berlin, Germany.,Institute of Molecular Tumor Biology, University of Münster, 48149, Münster, Germany
| | - Thomas K Albert
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Dennis Kastrati
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Amelie Alfert
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Till Holsten
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.,Institute of Molecular Tumor Biology, University of Münster, 48149, Münster, Germany
| | - Flavia de Faria
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany.,Department of Pediatric Hematology and Oncology, Children's Hospital of Brasìlia, 70684-831, Brasìlia, Brazil
| | - Michael Meisterernst
- Institute of Molecular Tumor Biology, University of Münster, 48149, Münster, Germany
| | - Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany
| | - Monika Warmuth-Metz
- Neuroradiological Reference Center, University Hospital Würzburg, Würzburg, Germany
| | - Johannes Nowak
- Neuroradiological Reference Center, University Hospital Würzburg, Würzburg, Germany.,SRH Poliklinik Gera GmbH, Radiological Practice Gotha, Gotha, Germany
| | - Gerd Meyer Zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, 48149, Münster, Germany
| | - Chloe Mayère
- Department of Genetic Medicine and Development, University of Geneva, 1211, Geneva, Switzerland.,iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211, Geneva, Switzerland
| | - Serge Nef
- Department of Genetic Medicine and Development, University of Geneva, 1211, Geneva, Switzerland.,iGE3, Institute of Genetics and Genomics of Geneva, University of Geneva, 1211, Geneva, Switzerland
| | - Pascal Johann
- Swabian Children's Cancer Center, Paediatric and Adolescent Medicine, University Medical Center Augsburg, 86156, Augsburg, Germany.,Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael C Frühwald
- Swabian Children's Cancer Center, Paediatric and Adolescent Medicine, University Medical Center Augsburg, 86156, Augsburg, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany.,Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Ulrich Schüller
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.,Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.,Research Institute Children's Cancer Center, 20251, Hamburg, Germany
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, 48149, Münster, Germany.
| |
Collapse
|
13
|
Li G, Wang C, Guan X, Bai Y, Feng Y, Wei W, Meng H, Fu M, He M, Zhang X, Lu Y, Lin Y, Guo H. Age-related DNA methylation on Y chromosome and their associations with total mortality among Chinese males. Aging Cell 2022; 21:e13563. [PMID: 35120273 PMCID: PMC8920452 DOI: 10.1111/acel.13563] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/10/2022] [Accepted: 01/24/2022] [Indexed: 11/28/2022] Open
Abstract
In view of the sex differences in aging‐related diseases, sex chromosomes may play a critical role during aging process. This study aimed to identify age‐related DNA methylation changes on Y chromosome (ChrY). A two‐stage study design was conducted in this study. The discovery stage contained 419 Chinese males, including 205 from the Wuhan‐Zhuhai cohort panel, 107 from the coke oven workers panel, and 107 from the Shiyan panel. The validation stage contained 587 Chinese males from the Dongfeng‐Tongji sub‐cohort. We used the Illumina HumanMethylation BeadChip to determine genome‐wide DNA methylation in peripheral blood of the study participants. The associations between age and methylation levels of ChrY CpGs were investigated by using linear regression models with adjustment for potential confounders. Further, associations of age‐related ChrY CpGs with all‐cause mortality were tested in the validation stage. We identified the significant associations of 41 ChrY CpGs with age at false discovery rate (FDR) <0.05 in the discovery stage, and 18 of them were validated in the validation stage (p < 0.05). Meta‐analysis of both stages confirmed the robust positive associations of 14 CpGs and negative associations of 4 CpGs with age (FDR<0.05). Among them, cg03441493 and cg17816615 were significantly associated with all‐cause mortality risk [HR(95% CI) = 1.37 (1.04, 1.79) and 0.70 (0.54, 0.93), respectively]. Our results highlighted the importance of ChrY CpGs on male aging.
Collapse
Affiliation(s)
- Guyanan Li
- Department of Occupational and Environmental Health State Key Laboratory of Environmental Health (Incubating) School of Public Health Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Department of Clinical Laboratory Medicine Shanghai Fifth People's Hospital Fudan University Shanghai China
| | - Chenming Wang
- Department of Occupational and Environmental Health State Key Laboratory of Environmental Health (Incubating) School of Public Health Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xin Guan
- Department of Occupational and Environmental Health State Key Laboratory of Environmental Health (Incubating) School of Public Health Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yansen Bai
- Department of Occupational and Environmental Health State Key Laboratory of Environmental Health (Incubating) School of Public Health Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yue Feng
- Department of Occupational and Environmental Health State Key Laboratory of Environmental Health (Incubating) School of Public Health Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Wei Wei
- Department of Occupational and Environmental Health State Key Laboratory of Environmental Health (Incubating) School of Public Health Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Hua Meng
- Department of Occupational and Environmental Health State Key Laboratory of Environmental Health (Incubating) School of Public Health Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ming Fu
- Department of Occupational and Environmental Health State Key Laboratory of Environmental Health (Incubating) School of Public Health Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Meian He
- Department of Occupational and Environmental Health State Key Laboratory of Environmental Health (Incubating) School of Public Health Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health State Key Laboratory of Environmental Health (Incubating) School of Public Health Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yanjun Lu
- Department of Laboratory Medicine Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yong Lin
- Department of Clinical Laboratory Medicine Shanghai Fifth People's Hospital Fudan University Shanghai China
- Department of Laboratory Medicine Huashan Hospital Fudan University Shanghai China
- National Clinical Research Center for Aging and Medicine Huashan Hospital Fudan University Shanghai China
| | - Huan Guo
- Department of Occupational and Environmental Health State Key Laboratory of Environmental Health (Incubating) School of Public Health Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
14
|
Guimarães Nunes IJ, Feltes BC, David MZ, Dorn M. Gene Expression Variation Analysis (GEVA): a new R package to evaluate variations in differential expression in multiple biological conditions. J Biomed Inform 2022; 129:104053. [DOI: 10.1016/j.jbi.2022.104053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/28/2022] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
|
15
|
Theocharidis G, Thomas BE, Sarkar D, Mumme HL, Pilcher WJR, Dwivedi B, Sandoval-Schaefer T, Sîrbulescu RF, Kafanas A, Mezghani I, Wang P, Lobao A, Vlachos IS, Dash B, Hsia HC, Horsley V, Bhasin SS, Veves A, Bhasin M. Single cell transcriptomic landscape of diabetic foot ulcers. Nat Commun 2022; 13:181. [PMID: 35013299 PMCID: PMC8748704 DOI: 10.1038/s41467-021-27801-8] [Citation(s) in RCA: 204] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
Diabetic foot ulceration (DFU) is a devastating complication of diabetes whose pathogenesis remains incompletely understood. Here, we profile 174,962 single cells from the foot, forearm, and peripheral blood mononuclear cells using single-cell RNA sequencing. Our analysis shows enrichment of a unique population of fibroblasts overexpressing MMP1, MMP3, MMP11, HIF1A, CHI3L1, and TNFAIP6 and increased M1 macrophage polarization in the DFU patients with healing wounds. Further, analysis of spatially separated samples from the same patient and spatial transcriptomics reveal preferential localization of these healing associated fibroblasts toward the wound bed as compared to the wound edge or unwounded skin. Spatial transcriptomics also validates our findings of higher abundance of M1 macrophages in healers and M2 macrophages in non-healers. Our analysis provides deep insights into the wound healing microenvironment, identifying cell types that could be critical in promoting DFU healing, and may inform novel therapeutic approaches for DFU treatment.
Collapse
Affiliation(s)
- Georgios Theocharidis
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics and Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Beena E Thomas
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - Debasree Sarkar
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - Hope L Mumme
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - William J R Pilcher
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - Bhakti Dwivedi
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | | - Ruxandra F Sîrbulescu
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Antonios Kafanas
- Lincoln County Hospital, Northern Lincolnshire and Goole NHS Foundation Trust, Scunthorpe, UK
| | - Ikram Mezghani
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics and Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Peng Wang
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics and Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Antonio Lobao
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics and Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Ioannis S Vlachos
- Department of Pathology, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, MA, USA
| | - Biraja Dash
- Yale Plastic and Reconstructive Surgery-Wound Center, Yale School of Medicine, New Haven, CT, USA
| | - Henry C Hsia
- Yale Plastic and Reconstructive Surgery-Wound Center, Yale School of Medicine, New Haven, CT, USA
| | - Valerie Horsley
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Swati S Bhasin
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - Aristidis Veves
- The Rongxiang Xu, MD, Center for Regenerative Therapeutics and Joslin-Beth Israel Deaconess Foot Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| | - Manoj Bhasin
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Department of Pediatrics and Biomedical Informatics, Emory University, Atlanta, GA, USA.
| |
Collapse
|
16
|
Taniue K, Tanu T, Shimoura Y, Mitsutomi S, Han H, Kakisaka R, Ono Y, Tamamura N, Takahashi K, Wada Y, Mizukami Y, Akimitsu N. RNA Exosome Component EXOSC4 Amplified in Multiple Cancer Types Is Required for the Cancer Cell Survival. Int J Mol Sci 2022; 23:496. [PMID: 35008922 PMCID: PMC8745236 DOI: 10.3390/ijms23010496] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/26/2021] [Accepted: 12/30/2021] [Indexed: 12/04/2022] Open
Abstract
The RNA exosome is a multi-subunit ribonuclease complex that is evolutionally conserved and the major cellular machinery for the surveillance, processing, degradation, and turnover of diverse RNAs essential for cell viability. Here we performed integrated genomic and clinicopathological analyses of 27 RNA exosome components across 32 tumor types using The Cancer Genome Atlas PanCancer Atlas Studies' datasets. We discovered that the EXOSC4 gene, which encodes a barrel component of the RNA exosome, was amplified across multiple cancer types. We further found that EXOSC4 alteration is associated with a poor prognosis of pancreatic cancer patients. Moreover, we demonstrated that EXOSC4 is required for the survival of pancreatic cancer cells. EXOSC4 also repressed BIK expression and destabilized SESN2 mRNA by promoting its degradation. Furthermore, knockdown of BIK and SESN2 could partially rescue pancreatic cells from the reduction in cell viability caused by EXOSC4 knockdown. Our study provides evidence for EXOSC4-mediated regulation of BIK and SESN2 mRNA in the survival of pancreatic tumor cells.
Collapse
Affiliation(s)
- Kenzui Taniue
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan; (T.T.); (Y.S.); (S.M.); (H.H.); (Y.W.)
- Cancer Genomics and Precision Medicine, Department of Medicine, Asahikawa Medical University, Asahikawa 078-8510, Japan; (N.T.); (K.T.); (Y.M.)
| | - Tanzina Tanu
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan; (T.T.); (Y.S.); (S.M.); (H.H.); (Y.W.)
| | - Yuki Shimoura
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan; (T.T.); (Y.S.); (S.M.); (H.H.); (Y.W.)
| | - Shuhei Mitsutomi
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan; (T.T.); (Y.S.); (S.M.); (H.H.); (Y.W.)
| | - Han Han
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan; (T.T.); (Y.S.); (S.M.); (H.H.); (Y.W.)
| | - Rika Kakisaka
- Institute of Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo 065-0033, Japan; (R.K.); (Y.O.)
| | - Yusuke Ono
- Institute of Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo 065-0033, Japan; (R.K.); (Y.O.)
| | - Nobue Tamamura
- Cancer Genomics and Precision Medicine, Department of Medicine, Asahikawa Medical University, Asahikawa 078-8510, Japan; (N.T.); (K.T.); (Y.M.)
| | - Kenji Takahashi
- Cancer Genomics and Precision Medicine, Department of Medicine, Asahikawa Medical University, Asahikawa 078-8510, Japan; (N.T.); (K.T.); (Y.M.)
| | - Youichiro Wada
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan; (T.T.); (Y.S.); (S.M.); (H.H.); (Y.W.)
| | - Yusuke Mizukami
- Cancer Genomics and Precision Medicine, Department of Medicine, Asahikawa Medical University, Asahikawa 078-8510, Japan; (N.T.); (K.T.); (Y.M.)
| | - Nobuyoshi Akimitsu
- Isotope Science Center, The University of Tokyo, Tokyo 113-0032, Japan; (T.T.); (Y.S.); (S.M.); (H.H.); (Y.W.)
| |
Collapse
|
17
|
Xicota L, De Toma I, Maffioletti E, Pisanu C, Squassina A, Baune BT, Potier MC, Stacey D, Dierssen M. Recommendations for pharmacotranscriptomic profiling of drug response in CNS disorders. Eur Neuropsychopharmacol 2022; 54:41-53. [PMID: 34743061 DOI: 10.1016/j.euroneuro.2021.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 08/16/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022]
Abstract
Pharmacotranscriptomics is a still very new field of research that has just begun to flourish and promises to enable target discovery, inform biomarker and evaluate drug efficacy beyond pharmacogenomics. The aim of this review is to provide a critical overview of the biological foundations of transcriptomics, methodological approaches to transcriptomic studies, and their advantages and limitations. We present the different RNA species (rRNAs, tRNAs, mtRNAs, snRNAs, scRNAs, mRNAs, ncRNAs, LINE and SINE transcripts, circular RNAs, piRNAs, miRNAs, snoRNAs) and their potential for pharmacotranscriptomic studies as markers to predict treatment response in neurological and psychiatric disorders. We also review the accessible sources of RNA in patients peripheral blood cells (including platelets), plasma, microvesicles, exosomes, apoptotic bodies, and how those affect the integrity and relative abundances of RNAs and reflect the situation in the Central Nervous System (CNS). Finally, we discuss the suitability and indications of different techniques, such as microarrays and RNA-sequencing (RNA-Seq) techniques to understand gene expression differences or to reveal variation in expression levels of coding and non-coding genes. We conclude with some recommendations for future directions, e.g., gaps of knowledge and particular RNAs/tissues that have been overlooked.
Collapse
Affiliation(s)
- Laura Xicota
- Paris Brain Institute, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Ilario De Toma
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Elisabetta Maffioletti
- Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Claudia Pisanu
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy
| | - Alessio Squassina
- Department of Biomedical Sciences, Section of Neuroscience and Clinical Pharmacology, University of Cagliari, Cagliari, Italy; Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Bernhard T Baune
- Department of Psychiatry, University of Muenster, Muenster, Germany; Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Marie Claude Potier
- Paris Brain Institute, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - David Stacey
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom of Great Britain and Northern Ireland United Kingdom
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Biomedical Research Networking Center on Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.
| | | |
Collapse
|
18
|
Epigenetic Regulation of Interleukin-17-Related Genes and Their Potential Roles in Neutrophil Vascular Infiltration in Preeclampsia. Reprod Sci 2022; 29:154-162. [PMID: 33959890 PMCID: PMC8571121 DOI: 10.1007/s43032-021-00605-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 05/03/2021] [Indexed: 01/03/2023]
Abstract
DNA methylation is an epigenetic mechanism controlling gene expression, and reduced methylation is associated with increased gene expression. We hypothesized that IL-17 cytokines are regulated by DNA methylation, are elevated in the circulation of preeclamptic women, and stimulate vascular neutrophil chemokine expression, which could account for vascular infiltration of neutrophils in preeclampsia. We found significantly reduced DNA methylation of IL17A, IL17E, and IL17F genes in omental arteries of preeclamptic women, significantly reduced methylation of IL2, which regulates IL-17-producing T-lymphocytes, and significantly reduced methylation of genes encoding neutrophil chemokines and TNFα receptors related to lymphocyte function. Maternal plasma levels of IL-17A were significantly elevated in the second trimester of preeclamptic pregnancy as compared to normal pregnancy. To test if methylation regulates IL-17 cytokines, a lymphocyte cell line (Jurkat) was cultured with a hypomethylating agent. Hypomethylation increased expression of IL17E (aka IL25), IL17F, and IL2. IL17A was not expressed by Jurkat cells. To test the potential role of IL-17 cytokines in vascular neutrophil infiltration associated with preeclampsia, human vascular smooth muscle cells were cultured with IL-17 cytokines. IL-17A, but not IL-17E or IL-17F, increased gene expression of neutrophil chemokines (IL-8, CXCL5, and CXCL6) that are increased in vascular smooth muscle of preeclamptic women. The monocyte chemokine, CCL-2, was not increased. TNFα also increased neutrophil chemokines. IL-17 cytokines are regulated by DNA methylation; IL-17A is elevated in preeclampsia and stimulates expression of neutrophil chemokines in vascular smooth muscle. IL-17A could be responsible for vascular infiltration of neutrophils in preeclampsia.
Collapse
|
19
|
Barata T, Vieira V, Rodrigues R, Neves RPD, Rocha M. Reconstruction of tissue-specific genome-scale metabolic models for human cancer stem cells. Comput Biol Med 2021; 142:105177. [PMID: 35026576 DOI: 10.1016/j.compbiomed.2021.105177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 02/07/2023]
Abstract
Cancer Stem Cells (CSCs) contribute to cancer aggressiveness, metastasis, chemo/radio-therapy resistance, and tumor recurrence. Recent studies emphasized the importance of metabolic reprogramming of CSCs for the maintenance and progression of the cancer phenotype through both the fulfillment of the energetic requirements and the supply of substrates fundamental for fast-cell growth, as well as through metabolite-induced epigenetic regulation. Therefore, it is of paramount importance to develop therapeutic strategies tailored to target the metabolism of CSCs. In this work, we built computational Genome-Scale Metabolic Models (GSMMs) for CSCs of different tissues. Flux simulations were then used to predict metabolic phenotypes, identify potential therapeutic targets, and spot already-known Transcription Factors (TFs), miRNAs and antimetabolites that could be used as part of drug repurposing strategies against cancer. Results were in accordance with experimental evidence, provided insights of new metabolic mechanisms for already known agents, and allowed for the identification of potential new targets and compounds that could be interesting for further in vitro and in vivo validation.
Collapse
Affiliation(s)
- Tânia Barata
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-517, Coimbra, Portugal
| | - Vítor Vieira
- Centre of Biological Engineering, University of Minho - Campus de Gualtar, Braga, Portugal
| | - Rúben Rodrigues
- Centre of Biological Engineering, University of Minho - Campus de Gualtar, Braga, Portugal
| | - Ricardo Pires das Neves
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-517, Coimbra, Portugal; IIIUC-Institute of Interdisciplinary Research, University of Coimbra, 3030-789, Coimbra, Portugal.
| | - Miguel Rocha
- Centre of Biological Engineering, University of Minho - Campus de Gualtar, Braga, Portugal; Department of Informatics, University of Minho.
| |
Collapse
|
20
|
Rodriguez-Gonzalez JC, Hernández-Balmaseda I, Declerck K, Pérez-Novo C, Logie E, Theys C, Jakubek P, Quiñones-Maza OL, Dantas-Cassali G, Carlos Dos Reis D, Van Camp G, Lopes Paz MT, Rodeiro-Guerra I, Delgado-Hernández R, Vanden Berghe W. Antiproliferative, Antiangiogenic, and Antimetastatic Therapy Response by Mangiferin in a Syngeneic Immunocompetent Colorectal Cancer Mouse Model Involves Changes in Mitochondrial Energy Metabolism. Front Pharmacol 2021; 12:670167. [PMID: 34924998 PMCID: PMC8678272 DOI: 10.3389/fphar.2021.670167] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 11/03/2021] [Indexed: 12/24/2022] Open
Abstract
In spite of the current advances and achievements in cancer treatments, colorectal cancer (CRC) persists as one of the most prevalent and deadly tumor types in both men and women worldwide. Drug resistance, adverse side effects and high rate of angiogenesis, metastasis and tumor relapse remain one of the greatest challenges in long-term management of CRC and urges need for new leads of anticancer drugs. We demonstrate that CRC treatment with the phytopharmaceutical mangiferin (MGF), a glucosylxanthone present in Mango tree stem bark and leaves (Mangifera Indica L.), induces dose-dependent tumor regression and decreases lung metastasis in a syngeneic immunocompetent allograft mouse model of murine CT26 colon carcinoma, which increases overall survival of mice. Antimetastatic and antiangiogenic MGF effects could be further validated in a wound healing in vitro model in human HT29 cells and in a matrigel plug implant mouse model. Interestingly, transcriptome pathway enrichment analysis demonstrates that MGF inhibits tumor growth, metastasis and angiogenesis by multi-targeting of mitochondrial oxidoreductase and fatty acid β-oxidation metabolism, PPAR, SIRT, NFκB, Stat3, HIF, Wnt and GP6 signaling pathways. MGF effects on fatty acid β-oxidation metabolism and carnitine palmitoyltransferase 1 (CPT1) protein expression could be further confirmed in vitro in human HT29 colon cells. In conclusion, antitumor, antiangiogenic and antimetastatic effects of MGF treatment hold promise to reduce adverse toxicity and to mitigate therapeutic outcome of colorectal cancer treatment by targeting mitochondrial energy metabolism in the tumor microenvironment.
Collapse
Affiliation(s)
| | | | - Ken Declerck
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Claudina Pérez-Novo
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Emilie Logie
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Claudia Theys
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| | - Patrycja Jakubek
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium.,Department of Food Chemistry, Technology and Biotechnology, Faculty of Chemistry, Gdansk University of Technology, Gdansk, Poland
| | | | - Geovanni Dantas-Cassali
- Departamento de Farmacología, Instituto de Ciencias Biológicas (ICB), Universidad Federal de Minas Gerais (UFMG), Horizonte, Brazil
| | - Diego Carlos Dos Reis
- Departamento de Farmacología, Instituto de Ciencias Biológicas (ICB), Universidad Federal de Minas Gerais (UFMG), Horizonte, Brazil
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Miriam Teresa Lopes Paz
- Departamento de Farmacología, Instituto de Ciencias Biológicas (ICB), Universidad Federal de Minas Gerais (UFMG), Horizonte, Brazil
| | - Idania Rodeiro-Guerra
- Laboratorio de Farmacología, Instituto de Ciencias del Mar (ICIMAR), CITMA, La Habana, Cuba
| | - René Delgado-Hernández
- Centro de Estudios para las Investigaciones y Evaluaciones Biológicas (CEIEB), Instituto de Farmacia y Alimentos (IFAL), Universidad de La Habana, La Habana, Cuba.,Facultad de Ciencias Naturales y Agropecuarias, Universidat de Santander (UDES), Bucaramanga, Colombia
| | - Wim Vanden Berghe
- Laboratory of Protein Science, Proteomics and Epigenetic Signaling (PPES) and Integrated Personalized and Precision Oncology Network (IPPON), Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, Antwerp, Belgium
| |
Collapse
|
21
|
Nunes IJG, Recamonde-Mendoza M, Feltes BC. Gene Expression Analysis Platform (GEAP): A highly customizable, fast, versatile and ready-to-use microarray analysis platform. Genet Mol Biol 2021; 45:e20210077. [PMID: 34927664 PMCID: PMC8754388 DOI: 10.1590/1678-4685-gmb-2021-0077] [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: 03/08/2021] [Accepted: 11/01/2021] [Indexed: 12/17/2022] Open
Abstract
There are still numerous challenges to be overcome in microarray data analysis because advanced, state-of-the-art analyses are restricted to programming users. Here we present the Gene Expression Analysis Platform, a versatile, customizable, optimized, and portable software developed for microarray analysis. GEAP was developed in C# for the graphical user interface, data querying, storage, results filtering and dynamic plotting, and R for data processing, quality analysis, and differential expression. Through a new automated system that identifies microarray file formats, retrieves contents, detects file corruption, and solves dependencies, GEAP deals with datasets independently of platform. GEAP covers 32 statistical options, supports quality assessment, differential expression from single and dual-channel experiments, and gene ontology. Users can explore results by different plots and filtering options. Finally, the entire data can be saved and organized through storage features, optimized for memory and data retrieval, with faster performance than R. These features, along with other new options, are not yet present in any microarray analysis software. GEAP accomplishes data analysis in a faster, straightforward, and friendlier way than other similar software, while keeping the flexibility for sophisticated procedures. By developing optimizations, unique customizations and new features, GEAP is destined for both advanced and non-programming users.
Collapse
Affiliation(s)
| | - Mariana Recamonde-Mendoza
- Universidade Federal do Rio Grande do Sul, Instituto de Informática, Porto Alegre, RS, Brazil.,Hospital de Clínicas de Porto Alegre (HCPA), Núcleo de Bioinformática, Porto Alegre, RS, Brazil
| | - Bruno César Feltes
- Universidade Federal do Rio Grande do Sul, Instituto de Informática, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Instituto de Biociências, Departamento de Genética, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul, Instituto de Biociências, Departamento de Biofísica, Porto Alegre, RS, Brazil
| |
Collapse
|
22
|
Patterns of Maternal Neutrophil Gene Expression at 30 Weeks of Gestation, but Not DNA Methylation, Distinguish Mild from Severe Preeclampsia. Int J Mol Sci 2021; 22:ijms222312876. [PMID: 34884685 PMCID: PMC8657979 DOI: 10.3390/ijms222312876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Neutrophils are activated and extensively infiltrate blood vessels in preeclamptic women. To identify genes that contribute to neutrophil activation and infiltration, we analyzed the transcriptomes of circulating neutrophils from normal pregnant and preeclamptic women. Neutrophils were collected at 30 weeks’ gestation and RNA and DNA were isolated for RNA sequencing and 5-hydroxy-methylcytosine (5-hmC) sequencing as an index of dynamic changes in neutrophil DNA methylation. Women with normal pregnancy who went on to develop mild preeclampsia at term had the most uniquely expressed genes (697) with 325 gene ontology pathways upregulated, many related to neutrophil activation and function. Women with severe preeclampsia who delivered prematurely had few pathways up- or downregulated. Cluster analysis revealed that gene expression in women with severe preeclampsia was an inverse mirror image of gene expression in normal pregnancy, while gene expression in women who developed mild preeclampsia was remarkably different from both. DNA methylation marks, key regulators of gene expression, are removed by the action of ten-eleven translocation (TET) enzymes, which oxidize 5-methylcytosines (5mCs), resulting in locus-specific reversal of DNA methylation. DNA sequencing for 5-hmC revealed no differences among the three groups. Genome-wide DNA methylation revealed extremely low levels in circulating neutrophils suggesting they are de-methylated. Collectively, these data demonstrate that neutrophil gene expression profiles can distinguish different preeclampsia phenotypes, and in the case of mild preeclampsia, alterations in gene expression occur well before clinical symptoms emerge. These findings serve as a foundation for further evaluation of neutrophil transcriptomes as biomarkers of preeclampsia phenotypes. Changes in DNA methylation in circulating neutrophils do not appear to mediate differential patterns of gene expression in either mild or severe preeclampsia.
Collapse
|
23
|
Ntunzwenimana JC, Boucher G, Paquette J, Gosselin H, Alikashani A, Morin N, Beauchamp C, Thauvette L, Rivard MÈ, Dupuis F, Deschênes S, Foisy S, Latour F, Lavallée G, Daly MJ, Xavier RJ, Charron G, Goyette P, Rioux JD. Functional screen of inflammatory bowel disease genes reveals key epithelial functions. Genome Med 2021; 13:181. [PMID: 34758847 PMCID: PMC8582123 DOI: 10.1186/s13073-021-00996-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/21/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Genetic studies have been tremendously successful in identifying genomic regions associated with a wide variety of phenotypes, although the success of these studies in identifying causal genes, their variants, and their functional impacts has been more limited. METHODS We identified 145 genes from IBD-associated genomic loci having endogenous expression within the intestinal epithelial cell compartment. We evaluated the impact of lentiviral transfer of the open reading frame (ORF) of these IBD genes into the HT-29 intestinal epithelial cell line via transcriptomic analyses. By comparing the genes in which expression was modulated by each ORF, as well as the functions enriched within these gene lists, we identified ORFs with shared impacts and their putative disease-relevant biological functions. RESULTS Analysis of the transcriptomic data for cell lines expressing the ORFs for known causal genes such as HNF4a, IFIH1, and SMAD3 identified functions consistent with what is already known for these genes. These analyses also identified two major clusters of genes: Cluster 1 contained the known IBD causal genes IFIH1, SBNO2, NFKB1, and NOD2, as well as genes from other IBD loci (ZFP36L1, IRF1, GIGYF1, OTUD3, AIRE and PITX1), whereas Cluster 2 contained the known causal gene KSR1 and implicated DUSP16 from another IBD locus. Our analyses highlight how multiple IBD gene candidates can impact on epithelial structure and function, including the protection of the mucosa from intestinal microbiota, and demonstrate that DUSP16 acts a regulator of MAPK activity and contributes to mucosal defense, in part via its regulation of the polymeric immunoglobulin receptor, involved in the protection of the intestinal mucosa from enteric microbiota. CONCLUSIONS This functional screen, based on expressing IBD genes within an appropriate cellular context, in this instance intestinal epithelial cells, resulted in changes to the cell's transcriptome that are relevant to their endogenous biological function(s). This not only helped in identifying likely causal genes within genetic loci but also provided insight into their biological functions. Furthermore, this work has highlighted the central role of intestinal epithelial cells in IBD pathophysiology, providing a scientific rationale for a drug development strategy that targets epithelial functions in addition to the current therapies targeting immune functions.
Collapse
Affiliation(s)
- Jessy Carol Ntunzwenimana
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Gabrielle Boucher
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Jean Paquette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Hugues Gosselin
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Azadeh Alikashani
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Nicolas Morin
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Claudine Beauchamp
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Louise Thauvette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Marie-Ève Rivard
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Frédérique Dupuis
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Sonia Deschênes
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Sylvain Foisy
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Frédéric Latour
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Geneviève Lavallée
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Mark J Daly
- Massachusetts General Hospital, Boston, MA, USA
- The Broad Institute, Cambridge, MA, USA
| | - Ramnik J Xavier
- Massachusetts General Hospital, Boston, MA, USA
- The Broad Institute, Cambridge, MA, USA
| | - Guy Charron
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Philippe Goyette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - John D Rioux
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada.
- Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
24
|
Fanidis D, Moulos P, Aidinis V. Fibromine is a multi-omics database and mining tool for target discovery in pulmonary fibrosis. Sci Rep 2021; 11:21712. [PMID: 34741074 PMCID: PMC8571330 DOI: 10.1038/s41598-021-01069-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/21/2021] [Indexed: 11/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a lethal lung fibroproliferative disease with limited therapeutic options. Differential expression profiling of affected sites has been instrumental for involved pathogenetic mechanisms dissection and therapeutic targets discovery. However, there have been limited efforts to comparatively analyse/mine the numerous related publicly available datasets, to fully exploit their potential on the validation/creation of novel research hypotheses. In this context and towards that goal, we present Fibromine, an integrated database and exploration environment comprising of consistently re-analysed, manually curated transcriptomic and proteomic pulmonary fibrosis datasets covering a wide range of experimental designs in both patients and animal models. Fibromine can be accessed via an R Shiny application (http://www.fibromine.com/Fibromine) which offers dynamic data exploration and real-time integration functionalities. Moreover, we introduce a novel benchmarking system based on transcriptomic datasets underlying characteristics, resulting to dataset accreditation aiming to aid the user on dataset selection. Cell specificity of gene expression can be visualised and/or explored in several scRNA-seq datasets, in an effort to link legacy data with this cutting-edge methodology and paving the way to their integration. Several use case examples are presented, that, importantly, can be reproduced on-the-fly by a non-specialist user, the primary target and potential user of this endeavour.
Collapse
Affiliation(s)
- Dionysios Fanidis
- Institute for Bioinnovation, Biomedical Sciences Research Center ″Alexander Fleming″, 16672, Athens, Greece
| | - Panagiotis Moulos
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center ″Alexander Fleming″, 16672, Athens, Greece.
| | - Vassilis Aidinis
- Institute for Bioinnovation, Biomedical Sciences Research Center ″Alexander Fleming″, 16672, Athens, Greece.
| |
Collapse
|
25
|
Lee MJ, Wang C, Carroll MJ, Brubaker DK, Hyman BT, Lauffenburger DA. Computational Interspecies Translation Between Alzheimer's Disease Mouse Models and Human Subjects Identifies Innate Immune Complement, TYROBP, and TAM Receptor Agonist Signatures, Distinct From Influences of Aging. Front Neurosci 2021; 15:727784. [PMID: 34658769 PMCID: PMC8515135 DOI: 10.3389/fnins.2021.727784] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/03/2021] [Indexed: 11/30/2022] Open
Abstract
Mouse models are vital for preclinical research on Alzheimer’s disease (AD) pathobiology. Many traditional models are driven by autosomal dominant mutations identified from early onset AD genetics whereas late onset and sporadic forms of the disease are predominant among human patients. Alongside ongoing experimental efforts to improve fidelity of mouse model representation of late onset AD, a computational framework termed Translatable Components Regression (TransComp-R) offers a complementary approach to leverage human and mouse datasets concurrently to enhance translation capabilities. We employ TransComp-R to integratively analyze transcriptomic data from human postmortem and traditional amyloid mouse model hippocampi to identify pathway-level signatures present in human patient samples yet predictive of mouse model disease status. This method allows concomitant evaluation of datasets across different species beyond observational seeking of direct commonalities between the species. Additional linear modeling focuses on decoupling disease signatures from effects of aging. Our results elucidated mouse-to-human translatable signatures associated with disease: excitatory synapses, inflammatory cytokine signaling, and complement cascade- and TYROBP-based innate immune activity; these signatures all find validation in previous literature. Additionally, we identified agonists of the Tyro3 / Axl / MerTK (TAM) receptor family as significant contributors to the cross-species innate immune signature; the mechanistic roles of the TAM receptor family in AD merit further dedicated study. We have demonstrated that TransComp-R can enhance translational understanding of relationships between AD mouse model data and human data, thus aiding generation of biological hypotheses concerning AD progression and holding promise for improved preclinical evaluation of therapies.
Collapse
Affiliation(s)
- Meelim J Lee
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Chuangqi Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Molly J Carroll
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Douglas K Brubaker
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States.,Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, IN, United States
| | - Bradley T Hyman
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Boston, MA, United States
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
26
|
Sieber-Frank J, Stark HJ, Kalteis S, Prigge ES, Köhler R, Andresen C, Henkel T, Casari G, Schubert T, Fischl W, Li-Weber M, Krammer PH, von Knebel Doeberitz M, Kopitz J, Kloor M, Ahadova A. Treatment resistance analysis reveals GLUT-1-mediated glucose uptake as a major target of synthetic rocaglates in cancer cells. Cancer Med 2021; 10:6807-6822. [PMID: 34546000 PMCID: PMC8495295 DOI: 10.1002/cam4.4212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 12/19/2022] Open
Abstract
Rocaglates are natural compounds that have been extensively studied for their ability to inhibit translation initiation. Rocaglates represent promising drug candidates for tumor treatment due to their growth‐inhibitory effects on neoplastic cells. In contrast to natural rocaglates, synthetic analogues of rocaglates have been less comprehensively characterized, but were also shown to have similar effects on the process of protein translation. Here, we demonstrate an enhanced growth‐inhibitory effect of synthetic rocaglates when combined with glucose anti‐metabolite 2‐deoxy‐D‐glucose (2DG) in different cancer cell lines. Moreover, we unravel a new aspect in the mechanism of action of synthetic rocaglates involving reduction of glucose uptake mediated by downregulation or abrogation of glucose transporter GLUT‐1 expression. Importantly, cells with genetically induced resistance to synthetic rocaglates showed substantially less pronounced treatment effect on glucose metabolism and did not demonstrate GLUT‐1 downregulation, pointing at the crucial role of this mechanism for the anti‐tumor activity of the synthetic rocaglates. Transcriptome profiling revealed glycolysis as one of the major pathways differentially regulated in sensitive and resistant cells. Analysis of synthetic rocaglate efficacy in a 3D tissue context with a co‐culture of tumor and normal cells demonstrated a selective effect on tumor cells and substantiated the mechanistic observations obtained in cancer cell lines. Increased glucose uptake and metabolism is a universal feature across different tumor types. Therefore, targeting this feature by synthetic rocaglates could represent a promising direction for exploitation of rocaglates in novel anti‐tumor therapies.
Collapse
Affiliation(s)
- Julia Sieber-Frank
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans-Jürgen Stark
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simon Kalteis
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elena-Sophie Prigge
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Richard Köhler
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carolin Andresen
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | | | | | - Min Li-Weber
- Tumor Immunology Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter H Krammer
- Tumor Immunology Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Magnus von Knebel Doeberitz
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Kopitz
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Aysel Ahadova
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
27
|
Edgar L, Akbar N, Braithwaite AT, Krausgruber T, Gallart-Ayala H, Bailey J, Corbin AL, Khoyratty TE, Chai JT, Alkhalil M, Rendeiro AF, Ziberna K, Arya R, Cahill TJ, Bock C, Laurencikiene J, Crabtree MJ, Lemieux ME, Riksen NP, Netea MG, Wheelock CE, Channon KM, Rydén M, Udalova IA, Carnicer R, Choudhury RP. Hyperglycemia Induces Trained Immunity in Macrophages and Their Precursors and Promotes Atherosclerosis. Circulation 2021; 144:961-982. [PMID: 34255973 PMCID: PMC8448412 DOI: 10.1161/circulationaha.120.046464] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/23/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Cardiovascular risk in diabetes remains elevated despite glucose-lowering therapies. We hypothesized that hyperglycemia induces trained immunity in macrophages, promoting persistent proatherogenic characteristics. METHODS Bone marrow-derived macrophages from control mice and mice with diabetes were grown in physiological glucose (5 mmol/L) and subjected to RNA sequencing (n=6), assay for transposase accessible chromatin sequencing (n=6), and chromatin immunoprecipitation sequencing (n=6) for determination of hyperglycemia-induced trained immunity. Bone marrow transplantation from mice with (n=9) or without (n=6) diabetes into (normoglycemic) Ldlr-/- mice was used to assess its functional significance in vivo. Evidence of hyperglycemia-induced trained immunity was sought in human peripheral blood mononuclear cells from patients with diabetes (n=8) compared with control subjects (n=16) and in human atherosclerotic plaque macrophages excised by laser capture microdissection. RESULTS In macrophages, high extracellular glucose promoted proinflammatory gene expression and proatherogenic functional characteristics through glycolysis-dependent mechanisms. Bone marrow-derived macrophages from diabetic mice retained these characteristics, even when cultured in physiological glucose, indicating hyperglycemia-induced trained immunity. Bone marrow transplantation from diabetic mice into (normoglycemic) Ldlr-/- mice increased aortic root atherosclerosis, confirming a disease-relevant and persistent form of trained innate immunity. Integrated assay for transposase accessible chromatin, chromatin immunoprecipitation, and RNA sequencing analyses of hematopoietic stem cells and bone marrow-derived macrophages revealed a proinflammatory priming effect in diabetes. The pattern of open chromatin implicated transcription factor Runt-related transcription factor 1 (Runx1). Similarly, transcriptomes of atherosclerotic plaque macrophages and peripheral leukocytes in patients with type 2 diabetes were enriched for Runx1 targets, consistent with a potential role in human disease. Pharmacological inhibition of Runx1 in vitro inhibited the trained phenotype. CONCLUSIONS Hyperglycemia-induced trained immunity may explain why targeting elevated glucose is ineffective in reducing macrovascular risk in diabetes and suggests new targets for disease prevention and therapy.
Collapse
Affiliation(s)
- Laurienne Edgar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | - Adam T. Braithwaite
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (T.K., A.F.R., C.B.)
| | - Héctor Gallart-Ayala
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.G.-A., C.E.W.)
- Department of Respiratory Medicine and Allergy (H.G.-A., C.E.W.), Karolinska University Hospital, Stockholm, Sweden
| | - Jade Bailey
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | - Alastair L. Corbin
- The Kennedy Institute of Rheumatology, University of Oxford, UK (A.L.C., T.E.K., I.A.U.)
| | - Tariq E. Khoyratty
- The Kennedy Institute of Rheumatology, University of Oxford, UK (A.L.C., T.E.K., I.A.U.)
| | - Joshua T. Chai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | - Mohammad Alkhalil
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | - André F. Rendeiro
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (T.K., A.F.R., C.B.)
| | - Klemen Ziberna
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | - Ritu Arya
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | - Thomas J. Cahill
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria (T.K., A.F.R., C.B.)
- Institute of Artificial Intelligence and Decision Support, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Austria (C.B.)
| | - Jurga Laurencikiene
- Department of Medicine (H7) (J.L., M.R.), Karolinska University Hospital, Stockholm, Sweden
| | - Mark J. Crabtree
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | | | - Niels P. Riksen
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands (N.P.R.., M.G.N.)
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands (N.P.R.., M.G.N.)
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Germany (M.G.N.)
| | - Craig E. Wheelock
- Division of Physiological Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden (H.G.-A., C.E.W.)
- Department of Respiratory Medicine and Allergy (H.G.-A., C.E.W.), Karolinska University Hospital, Stockholm, Sweden
| | - Keith M. Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | - Mikael Rydén
- Department of Medicine (H7) (J.L., M.R.), Karolinska University Hospital, Stockholm, Sweden
| | - Irina A. Udalova
- The Kennedy Institute of Rheumatology, University of Oxford, UK (A.L.C., T.E.K., I.A.U.)
| | - Ricardo Carnicer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| | - Robin P. Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK (L.E., N.A., A.T.B., J.B., J.T.C., M.A., K.Z., R.A., T.J.C., M.J.C., K.M.C., R.C., R.P.C.)
| |
Collapse
|
28
|
Yan Z, Wu Q, Cai W, Xiang H, Wen L, Zhang A, Peng Y, Zhang X, Wang H. Identifying critical genes associated with aneurysmal subarachnoid hemorrhage by weighted gene co-expression network analysis. Aging (Albany NY) 2021; 13:22345-22360. [PMID: 34542421 PMCID: PMC8507255 DOI: 10.18632/aging.203542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022]
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) is a life-threatening medical condition with a high mortality and disability rate. aSAH has an unclear pathogenesis, and limited treatment options are available. Here, we aimed to identify critical genes involved in aSAH pathogenesis using peripheral blood gene expression data of 43 patients with aSAH due to ruptured intracranial aneurysms and 18 controls with headache, downloaded from Gene Expression Omnibus. These data were used to construct a co-expression network using weighted gene co-expression network analysis (WGCNA). The biological functions of the hub genes were explored, and critical genes were selected by combining with differentially expressed genes analysis. Fourteen modules were identified by WGCNA. Among those modules, red, blue, brown and cyan modules were closely associated with aSAH. Moreover, 364 hub genes in the significant modules were found to play important roles in aSAH. Biological function analysis suggested that protein biosynthesis-related processes and inflammatory responses-related processes were involved in the pathology of aSAH pathology. Combined with differentially expressed genes analysis and validation in 35 clinical samples, seven gene (CD27, ANXA3, ACSL1, PGLYRP1, ALPL, ARG1, and TPST1) were identified as potential biomarkers for aSAH, and three genes (ANXA3, ALPL, and ARG1) were changed with disease development, that may provide new insights into potential molecular mechanisms for aSAH.
Collapse
Affiliation(s)
- Zhizhong Yan
- The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China.,Department of Neurosurgery, Jinling Hospital, Nanjing 210002, China.,Department of Neurosurgery, The 904th Hospital of The Joint Logistics Support Force of Chinese People's Liberation Army, Wuxi 214000, China
| | - Qi Wu
- Department of Neurosurgery, Jinling Hospital, Nanjing 210002, China
| | - Wei Cai
- Department of Neurosurgery, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian 223800, China
| | - Haitao Xiang
- Department of Neurosurgery, Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou 215028, China
| | - Lili Wen
- Department of Neurosurgery, Jinling Hospital, Nanjing 210002, China
| | - An Zhang
- Department of Neurosurgery, Jinling Hospital, Nanjing 210002, China
| | - Yaonan Peng
- Department of Neurosurgery, Jinling Hospital, Nanjing 210002, China
| | - Xin Zhang
- Department of Neurosurgery, Jinling Hospital, Nanjing 210002, China
| | - Handong Wang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China.,Department of Neurosurgery, Jinling Hospital, Nanjing 210002, China
| |
Collapse
|
29
|
Burel JG, Singhania A, Dubelko P, Muller J, Tanner R, Parizotto E, Dedicoat M, Fletcher TE, Dunbar J, Cunningham AF, Lindestam Arlehamn CS, Catanzaro DG, Catanzaro A, Rodwell T, McShane H, O'Shea MK, Peters B. Distinct blood transcriptomic signature of treatment in latent tuberculosis infected individuals at risk of developing active disease. Tuberculosis (Edinb) 2021; 131:102127. [PMID: 34555657 DOI: 10.1016/j.tube.2021.102127] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/28/2021] [Accepted: 09/09/2021] [Indexed: 01/12/2023]
Abstract
Although only a small fraction will ever develop the active form of tuberculosis (ATB) disease, chemoprophylaxis treatment in latent TB infected (LTBI) individuals is an effective strategy to control pathogen transmission. Characterizing immune responses in LTBI upon chemoprophylactic treatment is important to facilitate treatment monitoring, and thus improve TB control strategies. Here, we studied changes in the blood transcriptome in a cohort of 42 LTBI and 8 ATB participants who received anti-TB therapy. Based on the expression of previously published gene signatures of progression to ATB, we stratified the LTBI cohort in two groups and examined if individuals deemed to be at elevated risk of developing ATB before treatment (LTBI-Risk) differed from others (LTBI-Other). We found that LTBI-Risk and LTBI-Other groups were associated with two distinct transcriptomic treatment signatures, with the LTBI-Risk signature resembling that of treated ATB patients. Notably, overlapping genes between LTBI-Risk and ATB treatment signatures were associated with risk of progression to ATB and interferon (IFN) signaling, and were selectively downregulated upon treatment in the LTBI-Risk but not the LTBI-Other group. Our results suggest that transcriptomic reprogramming following treatment of LTBI is heterogeneous and can be used to distinguish LTBI-Risk individuals from the LTBI cohort at large.
Collapse
Affiliation(s)
- Julie G Burel
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Akul Singhania
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Paige Dubelko
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Julius Muller
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Rachel Tanner
- The Jenner Institute, University of Oxford, Oxford, UK
| | | | - Martin Dedicoat
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Thomas E Fletcher
- Royal Centre for Defence Medicine, Joint Medical Command, Birmingham, UK; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - James Dunbar
- Department of Infectious Diseases, The Friarage Hospital, Northallerton, UK
| | - Adam F Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | | | | | | - Timothy Rodwell
- Department of Medicine, University of California San Diego, CA, USA
| | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Matthew K O'Shea
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Royal Centre for Defence Medicine, Joint Medical Command, Birmingham, UK; Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| | - Bjoern Peters
- Vaccine Discovery Division, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Medicine, University of California San Diego, CA, USA.
| |
Collapse
|
30
|
Leal K, Saavedra K, Rebolledo C, Salazar LA. MicroRNAs hsa-miR-618 and hsa-miR-297 Might Modulate the Pleiotropic Effects Exerted by Statins in Endothelial Cells Through the Inhibition of ROCK2 Kinase: in-silico Approach. Front Cardiovasc Med 2021; 8:704175. [PMID: 34485404 PMCID: PMC8415262 DOI: 10.3389/fcvm.2021.704175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/20/2021] [Indexed: 01/11/2023] Open
Abstract
Several studies show that statin therapy improves endothelial function by cholesterol-independent mechanisms called “pleiotropic effects.” These are due to the inhibition of the RhoA/ROCK kinase pathway, its inhibition being an attractive atheroprotective treatment. In addition, recent work has shown that microRNAs, posttranscriptional regulators of gene expression, can affect the response of statins and their efficacy. For this reason, the objective of this study was to identify by bioinformatic analysis possible new microRNAs that could modulate the pleiotropic effects exerted by statins through the inhibition of ROCK kinases. A bioinformatic study was performed in which the differential expression of miRNAs in endothelial cells was compared under two conditions: Control and treated with simvastatin at 10 μM for 24 h, using a microarray. Seven miRNAs were differentially expressed, three up and four down. Within the up group, the miRNAs hsa-miR-618 and hsa-miR-297 present as a predicted target to ROCK2 kinase. Also, functional and enriched pathway analysis showed an association with mechanisms associated with atheroprotective effects. This work shows an in-silico approach of how posttranscriptional regulation mediated by miRNAs could modulate the pleiotropic effects exerted by statins on endothelial cells, through the inhibition of ROCK2 kinase and its effects.
Collapse
Affiliation(s)
- Karla Leal
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Kathleen Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Camilo Rebolledo
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Luis A Salazar
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
31
|
De Toma I, Sierra C, Dierssen M. Meta-analysis of transcriptomic data reveals clusters of consistently deregulated gene and disease ontologies in Down syndrome. PLoS Comput Biol 2021; 17:e1009317. [PMID: 34570756 PMCID: PMC8496798 DOI: 10.1371/journal.pcbi.1009317] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 10/07/2021] [Accepted: 07/31/2021] [Indexed: 01/04/2023] Open
Abstract
Trisomy of human chromosome 21 (HSA21) causes Down syndrome (DS). The trisomy does not simply result in the upregulation of HSA21--encoded genes but also leads to a genome-wide transcriptomic deregulation, which affect differently each tissue and cell type as a result of epigenetic mechanisms and protein-protein interactions. We performed a meta-analysis integrating the differential expression (DE) analyses of all publicly available transcriptomic datasets, both in human and mouse, comparing trisomic and euploid transcriptomes from different sources. We integrated all these data in a "DS network". We found that genome wide deregulation as a consequence of trisomy 21 is not arbitrary, but involves deregulation of specific molecular cascades in which both HSA21 genes and HSA21 interactors are more consistently deregulated compared to other genes. In fact, gene deregulation happens in "clusters", so that groups from 2 to 13 genes are found consistently deregulated. Most of these events of "co-deregulation" involve genes belonging to the same GO category, and genes associated with the same disease class. The most consistent changes are enriched in interferon related categories and neutrophil activation, reinforcing the concept that DS is an inflammatory disease. Our results also suggest that the impact of the trisomy might diverge in each tissue due to the different gene set deregulation, even though the triplicated genes are the same. Our original method to integrate transcriptomic data confirmed not only the importance of known genes, such as SOD1, but also detected new ones that could be extremely useful for generating or confirming hypotheses and supporting new putative therapeutic candidates. We created "metaDEA" an R package that uses our method to integrate every kind of transcriptomic data and therefore could be used with other complex disorders, such as cancer. We also created a user-friendly web application to query Ensembl gene IDs and retrieve all the information of their differential expression across the datasets.
Collapse
Affiliation(s)
- Ilario De Toma
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Cesar Sierra
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mara Dierssen
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Barcelona, Spain
| |
Collapse
|
32
|
Vega RB, Brouwers B, Parsons SA, Stephens NA, Pino MF, Hodges A, Yi F, Yu G, Pratley RE, Smith SR, Sparks LM. An improvement in skeletal muscle mitochondrial capacity with short-term aerobic training is associated with changes in Tribbles 1 expression. Physiol Rep 2021; 8:e14416. [PMID: 32562350 PMCID: PMC7305239 DOI: 10.14814/phy2.14416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/17/2020] [Accepted: 03/17/2020] [Indexed: 12/18/2022] Open
Abstract
Exercise training and physical activity are known to be associated with high mitochondrial content and oxidative capacity in skeletal muscle. Metabolic diseases including obesity and insulin resistance are associated with low mitochondrial capacity in skeletal muscle. Certain transcriptional factors such as PGC-1α are known to mediate the exercise response; however, the precise molecular mechanisms involved in the adaptation to exercise are not completely understood. We performed multiple measurements of mitochondrial capacity both in vivo and ex vivo in lean or overweight individuals before and after an 18-day aerobic exercise training regimen. These results were compared to lean, active individuals. Aerobic training in these individuals resulted in a marked increase in mitochondrial oxidative respiratory capacity without an appreciable increase in mitochondrial content. These adaptations were associated with robust transcriptome changes. This work also identifies the Tribbles pseudokinase 1, TRIB1, as a potential mediator of the exercise response in human skeletal muscle.
Collapse
Affiliation(s)
- Rick B Vega
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Bram Brouwers
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | | | | | - Maria F Pino
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Andrew Hodges
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Fanchao Yi
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Gongxin Yu
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | | | - Steven R Smith
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| |
Collapse
|
33
|
Declerck K, Novo CP, Grielens L, Van Camp G, Suter A, Vanden Berghe W. Echinacea purpurea (L.) Moench treatment of monocytes promotes tonic interferon signaling, increased innate immunity gene expression and DNA repeat hypermethylated silencing of endogenous retroviral sequences. BMC Complement Med Ther 2021; 21:141. [PMID: 33980308 PMCID: PMC8114977 DOI: 10.1186/s12906-021-03310-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Background Herbal remedies of Echinacea purpurea tinctures are widely used today to reduce common cold respiratory tract infections. Methods Transcriptome, epigenome and kinome profiling allowed a systems biology level characterisation of genomewide immunomodulatory effects of a standardized Echinacea purpurea (L.) Moench extract in THP1 monocytes. Results Gene expression and DNA methylation analysis revealed that Echinaforce® treatment triggers antiviral innate immunity pathways, involving tonic IFN signaling, activation of pattern recognition receptors, chemotaxis and immunometabolism. Furthermore, phosphopeptide based kinome activity profiling and pharmacological inhibitor experiments with filgotinib confirm a key role for Janus Kinase (JAK)-1 dependent gene expression changes in innate immune signaling. Finally, Echinaforce® treatment induces DNA hypermethylation at intergenic CpG, long/short interspersed nuclear DNA repeat elements (LINE, SINE) or long termininal DNA repeats (LTR). This changes transcription of flanking endogenous retroviral sequences (HERVs), involved in an evolutionary conserved (epi) genomic protective response against viral infections. Conclusions Altogether, our results suggest that Echinaforce® phytochemicals strengthen antiviral innate immunity through tonic IFN regulation of pattern recognition and chemokine gene expression and DNA repeat hypermethylated silencing of HERVs in monocytes. These results suggest that immunomodulation by Echinaforce® treatment holds promise to reduce symptoms and duration of infection episodes of common cold corona viruses (CoV), Severe Acute Respiratory Syndrome (SARS)-CoV, and new occurring strains such as SARS-CoV-2, with strongly impaired interferon (IFN) response and weak innate antiviral defense. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-021-03310-5.
Collapse
Affiliation(s)
- Ken Declerck
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Antwerp, Belgium
| | - Claudina Perez Novo
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Antwerp, Belgium
| | - Lisa Grielens
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Antwerp, Belgium
| | - Guy Van Camp
- Center of Medical Genetics, Department of Biomedical Sciences, University of Antwerp (UA) and University Hospital Antwerp (UZA), Antwerp, Belgium
| | | | - Wim Vanden Berghe
- Laboratory of Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp (UA), Antwerp, Belgium.
| |
Collapse
|
34
|
Kim K, Wang X, Ragonnaud E, Bodogai M, Illouz T, DeLuca M, McDevitt RA, Gusev F, Okun E, Rogaev E, Biragyn A. Therapeutic B-cell depletion reverses progression of Alzheimer's disease. Nat Commun 2021; 12:2185. [PMID: 33846335 PMCID: PMC8042032 DOI: 10.1038/s41467-021-22479-4] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/12/2021] [Indexed: 01/16/2023] Open
Abstract
The function of B cells in Alzheimer's disease (AD) is not fully understood. While immunoglobulins that target amyloid beta (Aβ) may interfere with plaque formation and hence progression of the disease, B cells may contribute beyond merely producing immunoglobulins. Here we show that AD is associated with accumulation of activated B cells in circulation, and with infiltration of B cells into the brain parenchyma, resulting in immunoglobulin deposits around Aβ plaques. Using three different murine transgenic models, we provide counterintuitive evidence that the AD progression requires B cells. Despite expression of the AD-fostering transgenes, the loss of B cells alone is sufficient to reduce Aβ plaque burden and disease-associated microglia. It reverses behavioral and memory deficits and restores TGFβ+ microglia, respectively. Moreover, therapeutic depletion of B cells at the onset of the disease retards AD progression in mice, suggesting that targeting B cells may also benefit AD patients.
Collapse
Affiliation(s)
- Ki Kim
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Xin Wang
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Emeline Ragonnaud
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Monica Bodogai
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Tomer Illouz
- The Mina and Everard Goodman faculty of Life Sciences, Ramat Gan, Israel
- The Gonda Brain Research Center, Bar Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer's disease research, Bar Ilan University, Ramat Gan, Israel
| | - Marisa DeLuca
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA
| | - Ross A McDevitt
- Mouse Phenotyping Unit, Comparative Medicine Section, National Institute on Aging, Baltimore, MD, USA
| | - Fedor Gusev
- Department of Genomics and Human Genetics, Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Eitan Okun
- The Mina and Everard Goodman faculty of Life Sciences, Ramat Gan, Israel
- The Gonda Brain Research Center, Bar Ilan University, Ramat Gan, Israel
- The Paul Feder Laboratory on Alzheimer's disease research, Bar Ilan University, Ramat Gan, Israel
| | - Evgeny Rogaev
- Department of Genomics and Human Genetics, Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
- Center for Genetics and Genetic Technologies, Faculty of Biology, Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, USA
- Sirius University of Science and Technology, Sochi, Russia
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Immunology and Molecular Biology, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
35
|
Shrestha S, Singhal S, Sens DA, Somji S, Davis BA, Guyer R, Breen S, Kalonick M, Garrett SH. Elevated glucose represses lysosomal and mTOR-related genes in renal epithelial cells composed of progenitor CD133+ cells. PLoS One 2021; 16:e0248241. [PMID: 33764985 PMCID: PMC7993790 DOI: 10.1371/journal.pone.0248241] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 02/23/2021] [Indexed: 12/16/2022] Open
Abstract
Hyperglycemia is one of the major health concern in many parts of the world. One of the serious complications of high glucose levels is diabetic nephropathy. The preliminary microarray study performed on primary human renal tubular epithelial (hRTE) cells exposed to high glucose levels showed a significant downregulation of mTOR as well as its associated genes as well as lysosomal genes. Based on this preliminary data, the expression of various lysosomal genes as well as mTOR and its associated genes were analyzed in hRTE cells exposed to 5.5, 7.5, 11 and 16 mM glucose. The results validated the microarray analysis, which showed a significant decrease in the mRNA as well as protein expression of the selected genes as the concentration of glucose increased. Co-localization of lysosomal marker, LAMP1 with mTOR showed lower expression of mTOR as the glucose concentration increased, suggesting decrease in mTOR activity. Although the mechanism by which glucose affects the regulation of lysosomal genes is not well known, our results suggest that high levels of glucose may lead to decrease in mTOR expression causing the cells to enter an anabolic state with subsequent downregulation of lysosomal genes.
Collapse
Affiliation(s)
- Swojani Shrestha
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Sandeep Singhal
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Donald A. Sens
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Seema Somji
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Bethany A. Davis
- Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Rachel Guyer
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Spencer Breen
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Matthew Kalonick
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Scott H. Garrett
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
- * E-mail:
| |
Collapse
|
36
|
Li CI, Zhang Y, Cieślik M, Wu YM, Xiao L, Cobain E, Tang MTC, Cao X, Porter P, Guenthoer J, Robinson DR, Chinnaiyan AM. Cancer Cell Intrinsic and Immunologic Phenotypes Determine Clinical Outcomes in Basal-like Breast Cancer. Clin Cancer Res 2021; 27:3079-3093. [PMID: 33753452 DOI: 10.1158/1078-0432.ccr-20-3890] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/28/2021] [Accepted: 03/16/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Basal-like breast cancer (BLBC) is a particularly aggressive intrinsic molecular subtype of breast cancer that lacks targeted therapies. There is also no clinically useful test to risk stratify patients with BLBC. We hypothesized that a transcriptome-based phenotypic characterization of BLBC tumors and their microenvironments may overcome these challenges. EXPERIMENTAL DESIGN We conducted a retrospective correlative genomic sequencing study using a matched pairs design with validation in five independent cohorts. The study was conducted on a large population-based prospective cohort of the major molecular subtypes of breast cancer conducted in the greater Seattle-Puget Sound metropolitan area. Cases consisted of women 20-69 years of age first diagnosed with invasive breast cancer identified through the population-based Surveillance Epidemiology and End Results program. Patients for this analysis (n = 949) were identified from the 1,408 patients with stage I-III triple-negative breast cancer [estrogen receptor-negative (ER-), progesterone receptor-negative (PR-), HER2-]. Of the 949 women, 248 developed a recurrence after their initial diagnosis. A matched set of 67 recurrent and nonrecurrent BLBC tumors was subjected to transcriptome sequencing. Through RNA sequencing of the matched sets of recurrent and nonrecurrent BLBC tumors, we aimed to identify prognostic phenotypes.To identify nonredundant and uncorrelated prognostic genes, we used an ensemble of variable selection algorithms, which resulted in a ranking of genes on the basis of their expected utility in classification. Using leave-one-out cross-validation, we trained a random forest classifier on the basis of the top 21 genes (BRAVO-DX). Validations were performed in five independent triple-negative or BLBC cohorts, and biomarker robustness and transferability were demonstrated by employing real-time PCR. RESULTS We found that cancer cell intrinsic and immunologic phenotypes are independent predictors of recurrence. By simultaneously interrogating the tumor and its microenvironment, we developed a compound risk model that stratified patients into low-, medium-, and high-risk groups, with a 14%/56%/74% chance of recurrence, respectively. Biologically, the primary tumors of patients who developed a recurrence had increased growth factor signaling and stem-like features, while nonrecurrent tumors showed high lymphocyte infiltration with clonal expansion of T and B cells, as well as antitumor polarization of macrophages. We validated our model in five independent cohorts, including three large cohorts, where BRAVO-DX was highly informative in identifying patients with disease recurrence [HR, 6.79 (95% confidence interval (CI), 1.89-24.37); HR, 3.45 (95% CI, 2.41-4.93); and HR, 1.69 (95% CI, 1.17-2.46)]. A smaller gene set focused on the tumor immunophenotype, BRAVO-IMMUNE, was highly prognostic in all five cohorts. CONCLUSIONS Together, these results indicate that phenotypic characteristics of BLBCs and their microenvironment are associated with recurrence-free survival and demonstrate the utility of intrinsic and extrinsic phenotypes as independent prognostic biomarkers in BLBC. Pending further evaluation and validation, our prognostic model has the potential to inform clinical decision-making for patients with BLBC as it identifies those at high risk of rapidly progressing on standard chemotherapy, as well as those who may benefit from alternative first-line therapies.
Collapse
Affiliation(s)
- Christopher I Li
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Marcin Cieślik
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Yi-Mi Wu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Lanbo Xiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Erin Cobain
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Mei-Tzu C Tang
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan
| | - Peggy Porter
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jamie Guenthoer
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Dan R Robinson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan.
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
37
|
Corrêa T, Feltes BC, Gonzalez EA, Baldo G, Matte U. Network Analysis Reveals Proteins Associated with Aortic Dilatation in Mucopolysaccharidoses. Interdiscip Sci 2021; 13:34-43. [PMID: 33475959 DOI: 10.1007/s12539-020-00406-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 11/25/2020] [Accepted: 12/03/2020] [Indexed: 06/12/2023]
Abstract
Mucopolysaccharidoses are caused by a deficiency of enzymes involved in the degradation of glycosaminoglycans. Heart diseases are a significant cause of morbidity and mortality in MPS patients, even in conditions in which enzyme replacement therapy is available. In this sense, cardiovascular manifestations, such as heart hypertrophy, cardiac function reduction, increased left ventricular chamber, and aortic dilatation, are among the most frequent. However, the downstream events which influence the heart dilatation process are unclear. Here, we employed systems biology tools together with transcriptomic data to investigate new elements that may be involved in aortic dilatation in Mucopolysaccharidoses syndrome. We identified candidate genes involved in biological processes related to inflammatory responses, deposition of collagen, and lipid accumulation in the cardiovascular system that may be involved in aortic dilatation in the Mucopolysaccharidoses I and VII. Furthermore, we investigated the molecular mechanisms of losartan treatment in Mucopolysaccharidoses I mice to underscore how this drug acts to prevent aortic dilation. Our data indicate that the association between the TGF-b signaling pathway, Fos, and Col1a1 proteins can play an essential role in aortic dilation's pathophysiology and its subsequent improvement by losartan treatment.
Collapse
Affiliation(s)
- Thiago Corrêa
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Porto Alegre, 90035-903, Brazil
- Postgraduation Program on Genetics and Molecular Biology, Federal University of Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Bruno César Feltes
- Institute of Informatics, Federal University of Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Esteban Alberto Gonzalez
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Porto Alegre, 90035-903, Brazil
- Postgraduation Program on Genetics and Molecular Biology, Federal University of Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Guilherme Baldo
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Porto Alegre, 90035-903, Brazil
- Postgraduation Program on Genetics and Molecular Biology, Federal University of Rio Grande Do Sul, Porto Alegre, RS, Brazil
| | - Ursula Matte
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Porto Alegre, 90035-903, Brazil.
- Postgraduation Program on Genetics and Molecular Biology, Federal University of Rio Grande Do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
38
|
Li YY, Zhang S, Wang H, Zhang SX, Xu T, Chen SW, Zhang Y, Chen Y. Identification of Crucial Genes and Pathways Associated with Atherosclerotic Plaque in Diabetic Patients. Pharmgenomics Pers Med 2021; 14:211-220. [PMID: 33568933 PMCID: PMC7869704 DOI: 10.2147/pgpm.s281705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/26/2020] [Indexed: 01/20/2023] Open
Abstract
Background Patients with diabetes have more calcification in atherosclerotic plaque and a higher occurrence of secondary cardiovascular events than patients without diabetes. The objective of this study was to identify crucial genes involved in the development of diabetic atherosclerotic plaque using a bioinformatics approach. Methods Microarray dataset GSE118481 was downloaded from the Gene Expression Omnibus (GEO) database; the dataset included 6 patients with diabetic atherosclerotic plaque (DBT) and 6 nondiabetic patients with atherosclerotic plaque (Ctrl). Differentially expressed genes (DEG) between the DBT and Ctrl groups were identified and then subjected to functional enrichment analysis. Based on the enriched pathways of DEGs, diabetic atherosclerotic plaque-related pathways were screened using the comparative toxicogenomics database (CTD). We then constructed a protein–protein interaction (PPI) network and transcription factor (TF)–miRNA–mRNA network. Results A total of 243 DEGs were obtained in the DBT group compared with the Ctrl group, including 85 up-regulated and 158 down-regulated DEGs. Functional enrichment analysis showed that up-regulated DEGs were mainly enriched in isoprenoid metabolic process, DNA-binding TF activity, and response to virus. Additionally, DEGs participating in the toll-like receptor signaling pathway were closely related to diabetes, carotid stenosis, and insulin resistance. The TF–miRNA–mRNA network showed that toll-like receptor 4 (TLR4), BCL2-like 11 (BCL2L11), and glutamate-cysteine ligase catalytic subunit (GCLC) were hub genes. Furthermore, TLR4 was regulated by TF signal transducer and activator of transcription 6 (STAT6); BCL2L11 was targeted by hsa-miR-24-3p; and GCLC was regulated by nuclear factor, erythroid 2 like 2 (NFE2L2). Conclusion Identification of hub genes and pathways increased our understanding of the molecular mechanisms underlying the atherosclerotic plaque in patients with or without diabetes. These crucial genes (TLR4, BC2L11, and GCLC) might function as molecular biomarkers for diabetic atherosclerotic plaque.
Collapse
Affiliation(s)
- Yuan-Yuan Li
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Sheng Zhang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Hua Wang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Shun-Xiao Zhang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Ting Xu
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Shu-Wen Chen
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Yan Zhang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Yue Chen
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| |
Collapse
|
39
|
De Toma I, Dierssen M. Network analysis of Down syndrome and SARS-CoV-2 identifies risk and protective factors for COVID-19. Sci Rep 2021; 11:1930. [PMID: 33479353 PMCID: PMC7820501 DOI: 10.1038/s41598-021-81451-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
SARS-CoV-2 infection has spread uncontrollably worldwide while it remains unknown how vulnerable populations, such as Down syndrome (DS) individuals are affected by the COVID-19 pandemic. Individuals with DS have more risk of infections with respiratory complications and present signs of auto-inflammation. They also present with multiple comorbidities that are associated with poorer COVID-19 prognosis in the general population. All this might place DS individuals at higher risk of SARS-CoV-2 infection or poorer clinical outcomes. In order to get insight into the interplay between DS genes and SARS-cov2 infection and pathogenesis we identified the genes associated with the molecular pathways involved in COVID-19 and the host proteins interacting with viral proteins from SARS-CoV-2. We then analyzed the overlaps of these genes with HSA21 genes, HSA21 interactors and other genes consistently differentially expressed in DS (using public transcriptomic datasets) and created a DS-SARS-CoV-2 network. We detected COVID-19 protective and risk factors among HSA21 genes and interactors and/or DS deregulated genes that might affect the susceptibility of individuals with DS both at the infection stage and in the progression to acute respiratory distress syndrome. Our analysis suggests that at the infection stage DS individuals might be more susceptible to infection due to triplication of TMPRSS2, that primes the viral S protein for entry in the host cells. However, as the anti-viral interferon I signaling is also upregulated in DS, this might increase the initial anti-viral response, inhibiting viral genome release, viral replication and viral assembly. In the second pro-inflammatory immunopathogenic phase of the infection, the prognosis for DS patients might worsen due to upregulation of inflammatory genes that might favor the typical cytokine storm of COVID-19. We also detected strong downregulation of the NLRP3 gene, critical for maintenance of homeostasis against pathogenic infections, possibly leading to bacterial infection complications.
Collapse
Affiliation(s)
- Ilario De Toma
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- Biomedical Research Networking Center On Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
40
|
Gatti F, Mia S, Hammarström C, Frerker N, Fosby B, Wang J, Pietka W, Sundnes O, Hol J, Kasprzycka M, Haraldsen G. Nuclear IL-33 restrains the early conversion of fibroblasts to an extracellular matrix-secreting phenotype. Sci Rep 2021; 11:108. [PMID: 33420328 PMCID: PMC7794291 DOI: 10.1038/s41598-020-80509-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 12/11/2020] [Indexed: 11/10/2022] Open
Abstract
Interleukin (IL)-33 is a cytokine that appears to mediate fibrosis by signaling via its receptor ST2 (IL-33R/IL1RL1). It is also, however, a protein that after synthesis is sorted to the cell nucleus, where it appears to affect chromatin folding. Here we describe a novel role for nuclear IL-33 in regulating the fibroblast phenotype in murine kidney fibrosis driven by unilateral ureteral obstruction. Transcriptional profiling of IL-33-deficient kidneys 24 h after ligation revealed enhanced expression of fibrogenic genes and enrichment of gene sets involved in extracellular matrix formation and remodeling. These changes relied on intracellular effects of IL-33, because they were not reproduced by treatment with a neutralizing antibody to IL-33 that prevents IL-33R/ST2L receptor signaling nor were they observed in IL-33R/ST2-deficient kidneys. To further explore the intracellular function of IL-33, we established transcription profiles of human fibroblasts, observing that knockdown of IL-33 skewed the transcription profile from an inflammatory towards a myofibroblast phenotype, reflected in higher levels of COL3A1, COL5A1 and transgelin protein, as well as lower expression levels of IL6, CXCL8, CLL7 and CCL8. In conclusion, our findings suggest that nuclear IL-33 in fibroblasts dampens the initial profibrotic response until persistent stimuli, as enforced by UUO, can override this protective mechanism.
Collapse
Affiliation(s)
- Francesca Gatti
- Department of Pathology, University of Oslo and Oslo University Hospital, PO Box 4950, 0424, Rikshospitalet, Norway
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Rikshospitalet, Norway
| | - Sobuj Mia
- Department of Pathology, University of Oslo and Oslo University Hospital, PO Box 4950, 0424, Rikshospitalet, Norway
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Rikshospitalet, Norway
| | - Clara Hammarström
- Department of Pathology, University of Oslo and Oslo University Hospital, PO Box 4950, 0424, Rikshospitalet, Norway
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Rikshospitalet, Norway
| | - Nadine Frerker
- Department of Pathology, University of Oslo and Oslo University Hospital, PO Box 4950, 0424, Rikshospitalet, Norway
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Rikshospitalet, Norway
| | - Bjarte Fosby
- Department of Surgery, University of Oslo and Oslo University Hospital, Rikshospitalet, Norway
| | - Junbai Wang
- Department of Pathology, University of Oslo and Oslo University Hospital, PO Box 4950, 0424, Rikshospitalet, Norway
| | - Wojciech Pietka
- Department of Pathology, University of Oslo and Oslo University Hospital, PO Box 4950, 0424, Rikshospitalet, Norway
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Rikshospitalet, Norway
| | - Olav Sundnes
- Department of Pathology, University of Oslo and Oslo University Hospital, PO Box 4950, 0424, Rikshospitalet, Norway
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Rikshospitalet, Norway
| | - Johanna Hol
- Department of Pathology, University of Oslo and Oslo University Hospital, PO Box 4950, 0424, Rikshospitalet, Norway
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Rikshospitalet, Norway
| | - Monika Kasprzycka
- Department of Pathology, University of Oslo and Oslo University Hospital, PO Box 4950, 0424, Rikshospitalet, Norway
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Rikshospitalet, Norway
| | - Guttorm Haraldsen
- Department of Pathology, University of Oslo and Oslo University Hospital, PO Box 4950, 0424, Rikshospitalet, Norway.
- K.G. Jebsen Inflammation Research Centre, University of Oslo and Oslo University Hospital, Rikshospitalet, Norway.
| |
Collapse
|
41
|
Joshi H, Vastrad B, Joshi N, Vastrad C, Tengli A, Kotturshetti I. Identification of Key Pathways and Genes in Obesity Using Bioinformatics Analysis and Molecular Docking Studies. Front Endocrinol (Lausanne) 2021; 12:628907. [PMID: 34248836 PMCID: PMC8264660 DOI: 10.3389/fendo.2021.628907] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
Obesity is an excess accumulation of body fat. Its progression rate has remained high in recent years. Therefore, the aim of this study was to diagnose important differentially expressed genes (DEGs) associated in its development, which may be used as novel biomarkers or potential therapeutic targets for obesity. The gene expression profile of E-MTAB-6728 was downloaded from the database. After screening DEGs in each ArrayExpress dataset, we further used the robust rank aggregation method to diagnose 876 significant DEGs including 438 up regulated and 438 down regulated genes. Functional enrichment analysis was performed. These DEGs were shown to be significantly enriched in different obesity related pathways and GO functions. Then protein-protein interaction network, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. The module analysis was performed based on the whole PPI network. We finally filtered out STAT3, CORO1C, SERPINH1, MVP, ITGB5, PCM1, SIRT1, EEF1G, PTEN and RPS2 hub genes. Hub genes were validated by ICH analysis, receiver operating curve (ROC) analysis and RT-PCR. Finally a molecular docking study was performed to find small drug molecules. The robust DEGs linked with the development of obesity were screened through the expression profile, and integrated bioinformatics analysis was conducted. Our study provides reliable molecular biomarkers for screening and diagnosis, prognosis as well as novel therapeutic targets for obesity.
Collapse
Affiliation(s)
- Harish Joshi
- Department of Endocrinology, Endocrine and Diabetes Care Center, Hubbali, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, India
| | - Nidhi Joshi
- Department of Medicine, Dr. D. Y. Patil Medical College, Kolhapur, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, India
- *Correspondence: Chanabasayya Vastrad,
| | - Anandkumar Tengli
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru, India
| | - Iranna Kotturshetti
- Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, India
| |
Collapse
|
42
|
Huang H, Yu X, Han X, Hao J, Zhao J, Bebek G, Bao S, Prayson RA, Khalil AM, Jankowsky E, Yu JS. Piwil1 Regulates Glioma Stem Cell Maintenance and Glioblastoma Progression. Cell Rep 2021; 34:108522. [PMID: 33406417 PMCID: PMC7837390 DOI: 10.1016/j.celrep.2020.108522] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 10/12/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
Piwi proteins are a subfamily of Argonaute proteins that maintain germ cells in eukaryotes. However, the role of their human homologs in cancer stem cells, and more broadly in cancer, is poorly understood. Here, we report that Piwi-like family members are overexpressed in glioblastoma (GBM), with Piwil1 (Hiwi) most frequently overexpressed (88%). Piwil1 is enriched in glioma stem-like cells (GSCs) to maintain self-renewal. Silencing Piwil1 in GSCs leads to global changes in gene expression resulting in cell-cycle arrest, senescence, or apoptosis. Piwil1 knockdown increases expression of the transcriptional co-regulator BTG2 and the E3-ubiquitin ligase FBXW7, leading to reduced c-Myc expression, as well as loss of expression of stem cell factors Olig2 and Nestin. Piwil1 regulates mRNA stability of BTG2, FBXW7, and CDKN1B. In animal models of GBM, Piwil1 knockdown suppresses tumor growth and promotes mouse survival. These findings support a role of Piwil1 in GSC maintenance and glioblastoma progression. Huang et al. find that Piwil1 protein is overexpressed in glioblastoma and glioma stem cells (GSCs). Piwil1 maintains GSC self-renewal and survival by regulating gene expression. Targeting Piwil1 extends survival in mouse models of glioblastoma. Piwil1 represents a therapeutic vulnerability.
Collapse
Affiliation(s)
- Haidong Huang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE60, Cleveland, OH 44195, USA
| | - Xingjiang Yu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE60, Cleveland, OH 44195, USA
| | - Xiangzi Han
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE60, Cleveland, OH 44195, USA
| | - Jing Hao
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE60, Cleveland, OH 44195, USA
| | - Jianjun Zhao
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE60, Cleveland, OH 44195, USA
| | - Gurkan Bebek
- Department of Nutrition, Center for Proteomics and Bioinformatics, Case Western Reserve University, 10900 Euclid Avenue, BRB 921, Cleveland, OH 44106, USA
| | - Shideng Bao
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE60, Cleveland, OH 44195, USA
| | - Richard A Prayson
- Department of Anatomic Pathology, The Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Ahmad M Khalil
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Eckhard Jankowsky
- Center for RNA Science and Therapeutics, Case Western Reserve University, 10900 Euclid Avenue, Wood Bldg. 137, Cleveland, OH 44106, USA
| | - Jennifer S Yu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NE60, Cleveland, OH 44195, USA; Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue, CA50, Cleveland, OH 44195, USA.
| |
Collapse
|
43
|
Feltes BC, Poloni JDF, Nunes IJG, Faria SS, Dorn M. Multi-Approach Bioinformatics Analysis of Curated Omics Data Provides a Gene Expression Panorama for Multiple Cancer Types. Front Genet 2020; 11:586602. [PMID: 33329726 PMCID: PMC7719697 DOI: 10.3389/fgene.2020.586602] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/09/2020] [Indexed: 12/19/2022] Open
Abstract
Studies describing the expression patterns and biomarkers for the tumoral process increase in number every year. The availability of new datasets, although essential, also creates a confusing landscape where common or critical mechanisms are obscured amidst the divergent and heterogeneous nature of such results. In this work, we manually curated the Gene Expression Omnibus using rigorous filtering criteria to select the most homogeneous and highest quality microarray and RNA-seq datasets from multiple types of cancer. By applying systems biology approaches, combined with machine learning analysis, we investigated possible frequently deregulated molecular mechanisms underlying the tumoral process. Our multi-approach analysis of 99 curated datasets, composed of 5,406 samples, revealed 47 differentially expressed genes in all analyzed cancer types, which were all in agreement with the validation using TCGA data. Results suggest that the tumoral process is more related to the overexpression of core deregulated machinery than the underexpression of a given gene set. Additionally, we identified gene expression similarities between different cancer types not described before and performed an overall survival analysis using 20 cancer types. Finally, we were able to suggest a core regulatory mechanism that could be frequently deregulated.
Collapse
Affiliation(s)
- Bruno César Feltes
- Laboratory of Structural Bioinformatics and Computational Biology, Institute of Informatics, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Joice de Faria Poloni
- Laboratory of Structural Bioinformatics and Computational Biology, Institute of Informatics, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Sara Socorro Faria
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Marcio Dorn
- Laboratory of Structural Bioinformatics and Computational Biology, Institute of Informatics, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Center of Biotechnology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- National Institute of Science and Technology - Forensic Science, Porto Alegre, Brazil
| |
Collapse
|
44
|
Giannoudis A, Malki MI, Rudraraju B, Mohhamed H, Menon S, Liloglou T, Ali S, Carroll JS, Palmieri C. Activating transcription factor-2 (ATF2) is a key determinant of resistance to endocrine treatment in an in vitro model of breast cancer. Breast Cancer Res 2020; 22:126. [PMID: 33198803 PMCID: PMC7667764 DOI: 10.1186/s13058-020-01359-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Activating transcription factor-2 (ATF2), a member of the leucine zipper family of DNA binding proteins, has been implicated as a tumour suppressor in breast cancer. However, its exact role in breast cancer endocrine resistance is still unclear. We have previously shown that silencing of ATF2 leads to a loss in the growth-inhibitory effects of tamoxifen in the oestrogen receptor (ER)-positive, tamoxifen-sensitive MCF7 cell line and highlighted that this multi-faceted transcription factor is key to the effects of tamoxifen in an endocrine sensitive model. In this work, we explored further the in vitro role of ATF2 in defining the resistance to endocrine treatment. MATERIALS AND METHODS We knocked down ATF2 in TAMR, LCC2 and LCC9 tamoxifen-resistant breast cancer cell lines as well as the parental tamoxifen sensitive MCF7 cell line and investigated the effects on growth, colony formation and cell migration. We also performed a microarray gene expression profiling (Illumina Human HT12_v4) to explore alterations in gene expression between MCF7 and TAMRs after ATF2 silencing and confirmed gene expression changes by quantitative RT-PCR. RESULTS By silencing ATF2, we observed a significant growth reduction of TAMR, LCC2 and LCC9 with no such effect observed with the parental MCF7 cells. ATF2 silencing was also associated with a significant inhibition of TAMR, LCC2 and LCC9 cell migration and colony formation. Interestingly, knockdown of ATF2 enhanced the levels of ER and ER-regulated genes, TFF1, GREB1, NCOA3 and PGR, in TAMR cells both at RNA and protein levels. Microarray gene expression identified a number of genes known to mediate tamoxifen resistance, to be differentially regulated by ATF2 in TAMR in relation to the parental MCF7 cells. Moreover, differential pathway analysis confirmed enhanced ER activity after ATF2 knockdown in TAMR cells. CONCLUSION These data demonstrate that ATF2 silencing may overcome endocrine resistance and highlights further the dual role of this transcription factor that can mediate endocrine sensitivity and resistance by modulating ER expression and activity.
Collapse
Affiliation(s)
- Athina Giannoudis
- Department of Molecular and Clinical Cancer Medicine, The Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK
| | - Mohammed Imad Malki
- Department of Molecular and Clinical Cancer Medicine, The Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Bharath Rudraraju
- Department of Molecular and Clinical Cancer Medicine, The Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, London, UK
| | - Hisham Mohhamed
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Knight Cancer Institute School of Medicine, Portland, USA
| | - Suraj Menon
- Cancer Research UK, Cambridge Research Institute, University of Cambridge, Cambridge, UK
| | - Triantafillos Liloglou
- Department of Molecular and Clinical Cancer Medicine, The Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK
| | - Simak Ali
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, London, UK
| | - Jason S Carroll
- Cancer Research UK, Cambridge Research Institute, University of Cambridge, Cambridge, UK
| | - Carlo Palmieri
- Department of Molecular and Clinical Cancer Medicine, The Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, L69 3GE, UK.
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
45
|
Faber H, Kurtoic D, Krishnamoorthy G, Weber P, Pütz B, Müller-Myhsok B, Weber F, Andlauer TFM. Gene Expression in Spontaneous Experimental Autoimmune Encephalomyelitis Is Linked to Human Multiple Sclerosis Risk Genes. Front Immunol 2020; 11:2165. [PMID: 33072080 PMCID: PMC7531036 DOI: 10.3389/fimmu.2020.02165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/10/2020] [Indexed: 12/23/2022] Open
Abstract
Recent genome-wide association studies have identified over 230 genetic risk loci for multiple sclerosis. Current experimental autoimmune encephalomyelitis (EAE) models requiring active induction of disease may not be optimally suited for the characterization of the function of these genes. We have thus used gene expression profiling to study whether spontaneous opticospinal EAE (OSE) or MOG-induced EAE mirrors the genetic contribution to the pathogenesis of multiple sclerosis more faithfully. To this end, we compared gene expression in OSE and MOG EAE models and analyzed the relationship of both models to human multiple sclerosis risk genes and T helper cell biology. We observed stronger gene expression changes and an involvement of more pathways of the adaptive immune system in OSE than MOG EAE. Furthermore, we demonstrated a more extensive enrichment of human MS risk genes among transcripts differentially expressed in OSE than was the case for MOG EAE. Transcripts differentially expressed only in diseased OSE mice but not in MOG EAE were significantly enriched for T helper cell-specific transcripts. These transcripts are part of immune-regulatory pathways. The activation of the adaptive immune system and the enrichment of both human multiple sclerosis risk genes and T helper cell-specific transcripts were also observed in OSE mice showing only mild disease signs. These expression changes may, therefore, be indicative of processes at disease onset. In summary, more human multiple sclerosis risk genes were differentially expressed in OSE than was observed for MOG EAE, especially in TH1 cells. When studying the functional role of multiple sclerosis risk genes and pathways during disease onset and their interactions with the environment, spontaneous OSE may thus show advantages over MOG-induced EAE.
Collapse
Affiliation(s)
- Hans Faber
- Max Planck Institute of Psychiatry, Munich, Germany
| | | | | | - Peter Weber
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Benno Pütz
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Bertram Müller-Myhsok
- Max Planck Institute of Psychiatry, Munich, Germany.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Frank Weber
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Till F M Andlauer
- Max Planck Institute of Psychiatry, Munich, Germany.,Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
46
|
De Boeck A, Ahn BY, D'Mello C, Lun X, Menon SV, Alshehri MM, Szulzewsky F, Shen Y, Khan L, Dang NH, Reichardt E, Goring KA, King J, Grisdale CJ, Grinshtein N, Hambardzumyan D, Reilly KM, Blough MD, Cairncross JG, Yong VW, Marra MA, Jones SJM, Kaplan DR, McCoy KD, Holland EC, Bose P, Chan JA, Robbins SM, Senger DL. Glioma-derived IL-33 orchestrates an inflammatory brain tumor microenvironment that accelerates glioma progression. Nat Commun 2020; 11:4997. [PMID: 33020472 PMCID: PMC7536425 DOI: 10.1038/s41467-020-18569-4] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
Despite a deeper molecular understanding, human glioblastoma remains one of the most treatment refractory and fatal cancers. It is known that the presence of macrophages and microglia impact glioblastoma tumorigenesis and prevent durable response. Herein we identify the dual function cytokine IL-33 as an orchestrator of the glioblastoma microenvironment that contributes to tumorigenesis. We find that IL-33 expression in a large subset of human glioma specimens and murine models correlates with increased tumor-associated macrophages/monocytes/microglia. In addition, nuclear and secreted functions of IL-33 regulate chemokines that collectively recruit and activate circulating and resident innate immune cells creating a pro-tumorigenic environment. Conversely, loss of nuclear IL-33 cripples recruitment, dramatically suppresses glioma growth, and increases survival. Our data supports the paradigm that recruitment and activation of immune cells, when instructed appropriately, offer a therapeutic strategy that switches the focus from the cancer cell alone to one that includes the normal host environment.
Collapse
Affiliation(s)
- Astrid De Boeck
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bo Young Ahn
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Charlotte D'Mello
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Xueqing Lun
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Shyam V Menon
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Mana M Alshehri
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Frank Szulzewsky
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Yaoqing Shen
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Lubaba Khan
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ngoc Ha Dang
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Elliott Reichardt
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kimberly-Ann Goring
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jennifer King
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Cameron J Grisdale
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Natalie Grinshtein
- Department of Molecular Genetics, University of Toronto and Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute and the Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Karlyne M Reilly
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Michael D Blough
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - J Gregory Cairncross
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - David R Kaplan
- Department of Molecular Genetics, University of Toronto and Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, ON, Canada
| | - Kathy D McCoy
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Eric C Holland
- Divison of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Pinaki Bose
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jennifer A Chan
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Pathology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Stephen M Robbins
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Donna L Senger
- Clark Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
47
|
Wang Z, Sun D, Chen YJ, Xie X, Shi Y, Tabar V, Brennan CW, Bale TA, Jayewickreme CD, Laks DR, Alcantara Llaguno S, Parada LF. Cell Lineage-Based Stratification for Glioblastoma. Cancer Cell 2020; 38:366-379.e8. [PMID: 32649888 PMCID: PMC7494533 DOI: 10.1016/j.ccell.2020.06.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/21/2020] [Accepted: 06/02/2020] [Indexed: 12/29/2022]
Abstract
Glioblastoma, the predominant adult malignant brain tumor, has been computationally classified into molecular subtypes whose functional relevance remains to be comprehensively established. Tumors from genetically engineered glioblastoma mouse models initiated by identical driver mutations in distinct cells of origin portray unique transcriptional profiles reflective of their respective lineage. Here, we identify corresponding transcriptional profiles in human glioblastoma and describe patient-derived xenografts with species-conserved subtype-discriminating functional properties. The oligodendrocyte lineage-associated glioblastoma subtype requires functional ERBB3 and harbors unique therapeutic sensitivities. These results highlight the importance of cell lineage in glioblastoma independent of driver mutations and provide a methodology for functional glioblastoma classification for future clinical investigations.
Collapse
Affiliation(s)
- Zilai Wang
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Daochun Sun
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yu-Jung Chen
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xuanhua Xie
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yufeng Shi
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Viviane Tabar
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Cameron W Brennan
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tejus A Bale
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chenura D Jayewickreme
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Dan R Laks
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sheila Alcantara Llaguno
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Luis F Parada
- Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
48
|
Kolberg L, Kerimov N, Peterson H, Alasoo K. Co-expression analysis reveals interpretable gene modules controlled by trans-acting genetic variants. eLife 2020; 9:e58705. [PMID: 32880574 PMCID: PMC7470823 DOI: 10.7554/elife.58705] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022] Open
Abstract
Understanding the causal processes that contribute to disease onset and progression is essential for developing novel therapies. Although trans-acting expression quantitative trait loci (trans-eQTLs) can directly reveal cellular processes modulated by disease variants, detecting trans-eQTLs remains challenging due to their small effect sizes. Here, we analysed gene expression and genotype data from six blood cell types from 226 to 710 individuals. We used co-expression modules inferred from gene expression data with five methods as traits in trans-eQTL analysis to limit multiple testing and improve interpretability. In addition to replicating three established associations, we discovered a novel trans-eQTL near SLC39A8 regulating a module of metallothionein genes in LPS-stimulated monocytes. Interestingly, this effect was mediated by a transient cis-eQTL present only in early LPS response and lost before the trans effect appeared. Our analyses highlight how co-expression combined with functional enrichment analysis improves the identification and prioritisation of trans-eQTLs when applied to emerging cell-type-specific datasets.
Collapse
Affiliation(s)
- Liis Kolberg
- Institute of Computer Science, University of TartuTartuEstonia
| | - Nurlan Kerimov
- Institute of Computer Science, University of TartuTartuEstonia
| | - Hedi Peterson
- Institute of Computer Science, University of TartuTartuEstonia
| | - Kaur Alasoo
- Institute of Computer Science, University of TartuTartuEstonia
| |
Collapse
|
49
|
Gabriel M, Fey V, Heinosalo T, Adhikari P, Rytkönen K, Komulainen T, Huhtinen K, Laajala TD, Siitari H, Virkki A, Suvitie P, Kujari H, Aittokallio T, Perheentupa A, Poutanen M. A relational database to identify differentially expressed genes in the endometrium and endometriosis lesions. Sci Data 2020; 7:284. [PMID: 32859947 PMCID: PMC7455745 DOI: 10.1038/s41597-020-00623-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 08/03/2020] [Indexed: 01/06/2023] Open
Abstract
Endometriosis is a common inflammatory estrogen-dependent gynecological disorder, associated with pelvic pain and reduced fertility in women. Several aspects of this disorder and its cellular and molecular etiology remain unresolved. We have analyzed the global gene expression patterns in the endometrium, peritoneum and in endometriosis lesions of endometriosis patients and in the endometrium and peritoneum of healthy women. In this report, we present the EndometDB, an interactive web-based user interface for browsing the gene expression database of collected samples without the need for computational skills. The EndometDB incorporates the expression data from 115 patients and 53 controls, with over 24000 genes and clinical features, such as their age, disease stages, hormonal medication, menstrual cycle phase, and the different endometriosis lesion types. Using the web-tool, the end-user can easily generate various plot outputs and projections, including boxplots, and heatmaps and the generated outputs can be downloaded in pdf-format. Availability and implementationThe web-based user interface is implemented using HTML5, JavaScript, CSS, Plotly and R. It is freely available from https://endometdb.utu.fi/. Measurement(s) | RNA • differential expression analysis data • endometriosis | Technology Type(s) | Microarray Analysis • digital curation | Factor Type(s) | age • disease stage • menstrual cycle phase • hormonal medication • endometriosis lesion type • endometrium | Sample Characteristic - Organism | Homo sapiens |
Machine-accessible metadata file describing the reported data: 10.6084/m9.figshare.12800642
Collapse
Affiliation(s)
- Michael Gabriel
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, University of Turku, 20520, Turku, Finland.,Department of Obstetrics and Gynecology, University of Turku, and Turku University Hospital, 20014, Turku, Finland
| | - Vidal Fey
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, University of Turku, 20520, Turku, Finland
| | - Taija Heinosalo
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, University of Turku, 20520, Turku, Finland
| | - Prem Adhikari
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, University of Turku, 20520, Turku, Finland
| | - Kalle Rytkönen
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, University of Turku, 20520, Turku, Finland.,Turku Bioscience Centre, University of Turku and Åbo Akademi, Turku, Finland
| | - Tuomo Komulainen
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, University of Turku, 20520, Turku, Finland
| | - Kaisa Huhtinen
- Institute of Biomedicine, Research Center for Cancer, Infections and Immunity, University of Turku, 20520, Turku, Finland.,Department of Pathology, Turku University Hospital, 20521, Turku, Finland
| | - Teemu Daniel Laajala
- Department of Mathematics and Statistics, University of Turku, 20014, Turku, Finland.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Harri Siitari
- Department of Neurology, Faculty of Medicine, University of Turku, 20014, Turku, Finland
| | - Arho Virkki
- Department of Mathematics and Statistics, University of Turku, 20014, Turku, Finland
| | - Pia Suvitie
- Department of Obstetrics and Gynecology, University of Turku, and Turku University Hospital, 20014, Turku, Finland
| | - Harry Kujari
- Institute of Biomedicine, Research Center for Cancer, Infections and Immunity, University of Turku, 20520, Turku, Finland.,Department of Pathology, Turku University Hospital, 20521, Turku, Finland
| | - Tero Aittokallio
- Department of Mathematics and Statistics, University of Turku, 20014, Turku, Finland. .,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland.
| | - Antti Perheentupa
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, University of Turku, 20520, Turku, Finland. .,Department of Obstetrics and Gynecology, University of Turku, and Turku University Hospital, 20014, Turku, Finland.
| | - Matti Poutanen
- Institute of Biomedicine, Research Center for Integrative Physiology and Pharmacology, University of Turku, 20520, Turku, Finland. .,Institute of Medicine, Sahlgrenska Academy, 405 30 Gothenburg University, Gothenburg, Sweden.
| |
Collapse
|
50
|
Schettini F, Sobhani N, Ianza A, Triulzi T, Molteni A, Lazzari MC, Strina C, Milani M, Corona SP, Sirico M, Bernocchi O, Giudici F, Cappelletti MR, Ciruelos E, Jerusalem G, Loi S, Fox SB, Generali D. Immune system and angiogenesis-related potential surrogate biomarkers of response to everolimus-based treatment in hormone receptor-positive breast cancer: an exploratory study. Breast Cancer Res Treat 2020; 184:421-431. [PMID: 32770287 PMCID: PMC7599144 DOI: 10.1007/s10549-020-05856-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/31/2020] [Indexed: 01/08/2023]
Abstract
Purpose mTOR inhibitor everolimus is used for hormone receptor-positive (HR+)/HER2-negative metastatic breast cancer (mBC). No reliable predictive biomarker of response is available. Following evidences from other solid tumors, we aimed to assess the association between treatment-associated immune system features and everolimus activity. Methods We retrospectively explored a correlation with the therapeutic activity of everolimus and tumor-associated immune pathways with ingenuity pathway analysis (IPA), neutrophil-to-lymphocyte ratio (NLR), circulating lymphocytes, and endothelial cells (CECs) in 3 different HR+ mBC studies, including the BALLET phase IIIb study. Results The circulating levels of CD3+/CD8+, CD3+/CD4+, and overall T lymphocytes were higher in responders versus non-responders at baseline (p = 0.017, p < 0.001, p = 0.034) and after treatment (p = 0.01, p = 0.003, p = 0.023). Reduced CECs, a tumor neoangiogenesis marker, were observed in responders after treatment (p < 0.001). Patients with low NLR (≤ 4.4) showed a better progression-free survival compared to patients with high NLR (> 4.4) (p = 0.01). IPA showed that the majority of immunity-related genes were found upregulated in responders compared to non-responders before treatment, but not after. Conclusions Lymphocytes subpopulations, CECs and NLR could be interesting biomarkers predictive of response to everolimus-based regimens, potentially useful in daily clinical practice to select/monitor everolimus-based treatment in mBC. Further studies to confirm such hypotheses are warranted.
Collapse
Affiliation(s)
- Francesco Schettini
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy.,Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,SOLTI Breast Cancer Research Group, Barcelona, Spain
| | - Navid Sobhani
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Anna Ianza
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alfredo Molteni
- UO Ematologia e CTMO, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | | | - Carla Strina
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Manuela Milani
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Silvia Paola Corona
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Marianna Sirico
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Ottavia Bernocchi
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy.,UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Fabiola Giudici
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy
| | - Maria Rosaria Cappelletti
- UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Eva Ciruelos
- SOLTI Breast Cancer Research Group, Barcelona, Spain.,Department of Medical Oncology, Breast Cancer Unit, University Hospital, 12 de Octubre, Avda de Córdoba s/n, Madrid, Spain
| | - Guy Jerusalem
- Department of Medical Oncology, Centre Hospitalier Universitaire de Liège and Liège University, Avenue de L'Hòpital 1, 4000, Liège, Belgium
| | - Sherine Loi
- Peter MacCallum Cancer Center, 305 Grattan Street, Melbourne, VIC, Australia.,University of Melbourne, Melbourne, VIC, Australia
| | - Stephen B Fox
- Peter MacCallum Cancer Center, 305 Grattan Street, Melbourne, VIC, Australia.,University of Melbourne, Melbourne, VIC, Australia
| | - Daniele Generali
- Department of Medical, Surgery & Health Sciences, University of Trieste, Piazza Ospitale 1, 34129, Trieste, Italy. .,UO Multidisciplinare di Patologia Mammaria e Ricerca Traslazionale, ASST di Cremona, Viale Concordia 1, 26100, Cremona, Italy.
| |
Collapse
|