1
|
Liu Z, Wang H, Liang Y, Liu M, Huang Q, Wang M, Zhou J, Bu Q, Zhou H, Lu L. E2F2 Reprograms Macrophage Function By Modulating Material and Energy Metabolism in the Progression of Metabolic Dysfunction-Associated Steatohepatitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2410880. [PMID: 39465673 PMCID: PMC11672278 DOI: 10.1002/advs.202410880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/30/2024] [Indexed: 10/29/2024]
Abstract
Macrophages are essential for the development of steatosis, hepatic inflammation, and fibrosis in metabolic dysfunction-associated steatohepatitis(MASH). However, the roles of macrophage E2F2 in the progression of MASH have not been elucidated. This study reveals that the expression of macrophage E2F2 is dramatically downregulated in MASH livers from mice and humans, and that this expression is adversely correlated with the severity of the disease. Myeloid-specific E2F2 depletion aggravates intrahepatic inflammation, hepatic stellate cell activation, and hepatocyte lipid accumulation during MASH progression. Mechanistically, E2F2 can inhibit the SLC7A5 transcription directly. E2F2 deficiency upregulates the expression of SLC7A5 to mediate amino acids flux, resulting in enhanced glycolysis, impaired mitochondrial function, and increased macrophages proinflammatory response in a Leu-mTORC1-dependent manner. Moreover, bioinformatics analysis and CUT &Tag assay identify the direct binding of Nrf2 to E2F2 promoter to promote its transcription and nuclear translocation. Genetic or pharmacological activation of Nrf2 effectively activates E2F2 to attenuate the MASH progression. Finally, patients treated with CDK4/6 inhibitors demonstrate reduced E2F2 activity but increased SLC7A5 activity in PBMCs. These findings indicated macrophage E2F2 suppresses MASH progression by reprogramming amino acid metabolism via SLC7A5- Leu-mTORC1 signaling pathway. Activating E2F2 holds promise as a therapeutic strategy for MASH.
Collapse
Affiliation(s)
- Zheng Liu
- Department of General Surgerythe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029China
| | - Hao Wang
- Department of Liver SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100730China
| | - Yuan Liang
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
- School of Biological Science & Medical EngineeringSoutheast UniversityNanjing210096China
| | - Mu Liu
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Qiyuan Huang
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Mingming Wang
- Department of Liver SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100730China
| | - Jinren Zhou
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Qingfa Bu
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Haoming Zhou
- Hepatobiliary CenterThe First Affiliated Hospital of Nanjing Medical University& Research Unit of Liver Transplantation and Transplant ImmunologyChinese Academy of Medical SciencesNanjing210029China
| | - Ling Lu
- Affiliated Hospital of Xuzhou Medical UniversityXuzhou220005China
| |
Collapse
|
2
|
To B, Broeker C, Jhan JR, Garcia-Lerena J, Vusich J, Rempel R, Rennhack JP, Hollern D, Jackson L, Judah D, Swiatnicki M, Bylett E, Kubiak R, Honeysett J, Nevins J, Andrechek E. Insight into mammary gland development and tumor progression in an E2F5 conditional knockout mouse model. Oncogene 2024; 43:3402-3415. [PMID: 39341991 PMCID: PMC11554565 DOI: 10.1038/s41388-024-03172-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 08/23/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024]
Abstract
Development of breast cancer is linked to altered regulation of mammary gland developmental processes. A better understanding of normal mammary gland development can thus reveal possible mechanisms of how normal cells are re-programmed to become malignant. E2Fs 1-4 are part of the E2F transcription factor family with varied roles in mammary development, but little is known about the role of E2F5. A combination of scRNAseq and predictive signature tools demonstrated the presence of E2F5 in the mammary gland and showed changes in predicted activity during the various phases of mammary gland development. Testing the hypothesis that E2F5 regulates mammary function, we generated a mammary-specific E2F5 knockout mouse model, resulting in modest mammary gland development changes. However, after a prolonged latency the E2F5 conditional knockout mice developed highly metastatic mammary tumors. Whole genome sequencing revealed significant intertumor heterogeneity. RNAseq and protein analysis identified altered levels of Cyclin D1, with similarities to MMTV-Neu tumors, suggesting that E2F5 conditional knockout mammary glands and tumors may be dependent on Cyclin D1. Transplantation of the tumors revealed metastases to lymph nodes that were enriched through serial transplantation in immune competent recipients. Based on these findings, we propose that loss of E2F5 leads to altered regulation of Cyclin D1, which facilitates the development of metastatic mammary tumors after long latency. More importantly, this study demonstrates that conditional loss of E2F5 in the mammary gland leads to tumor formation, revealing its role as a transcription factor regulating a network of genes that normally result in a tumor suppressor function.
Collapse
Affiliation(s)
- Briana To
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Carson Broeker
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Jing-Ru Jhan
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | | | - John Vusich
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | | | | | | | - Lauren Jackson
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - David Judah
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Matt Swiatnicki
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Evan Bylett
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Rachel Kubiak
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Jordan Honeysett
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | | | - Eran Andrechek
- Department of Physiology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
3
|
Sankaran DG, Zhu H, Maymi VI, Forlastro IM, Jiang Y, Laniewski N, Scheible KM, Rudd BD, Grimson AW. Gene Regulatory Programs that Specify Age-Related Differences during Thymocyte Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.599011. [PMID: 38948840 PMCID: PMC11212896 DOI: 10.1101/2024.06.14.599011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
T cell development is fundamental to immune system establishment, yet how this development changes with age remains poorly understood. Here, we construct a transcriptional and epigenetic atlas of T cell developmental programs in neonatal and adult mice, revealing the ontogeny of divergent gene regulatory programs and their link to age-related differences in phenotype and function. Specifically, we identify a gene module that diverges with age from the earliest stages of genesis and includes programs that govern effector response and cell cycle regulation. Moreover, we reveal that neonates possess more accessible chromatin during early thymocyte development, likely establishing poised gene expression programs that manifest later in thymocyte development. Finally, we leverage this atlas, employing a CRISPR-based perturbation approach coupled with single-cell RNA sequencing as a readout to uncover a conserved transcriptional regulator, Zbtb20, that contributes to age-dependent differences in T cell development. Altogether, our study defines transcriptional and epigenetic programs that regulate age-specific differences in T cell development.
Collapse
|
4
|
von Hofsten S, Fenton KA, Pedersen HL. Human and Murine Toll-like Receptor-Driven Disease in Systemic Lupus Erythematosus. Int J Mol Sci 2024; 25:5351. [PMID: 38791389 PMCID: PMC11120885 DOI: 10.3390/ijms25105351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 05/26/2024] Open
Abstract
The pathogenesis of systemic lupus erythematosus (SLE) is linked to the differential roles of toll-like receptors (TLRs), particularly TLR7, TLR8, and TLR9. TLR7 overexpression or gene duplication, as seen with the Y-linked autoimmune accelerator (Yaa) locus or TLR7 agonist imiquimod, correlates with increased SLE severity, and specific TLR7 polymorphisms and gain-of-function variants are associated with enhanced SLE susceptibility and severity. In addition, the X-chromosome location of TLR7 and its escape from X-chromosome inactivation provide a genetic basis for female predominance in SLE. The absence of TLR8 and TLR9 have been shown to exacerbate the detrimental effects of TLR7, leading to upregulated TLR7 activity and increased disease severity in mouse models of SLE. The regulatory functions of TLR8 and TLR9 have been proposed to involve competition for the endosomal trafficking chaperone UNC93B1. However, recent evidence implies more direct, regulatory functions of TLR9 on TLR7 activity. The association between age-associated B cells (ABCs) and autoantibody production positions these cells as potential targets for treatment in SLE, but the lack of specific markers necessitates further research for precise therapeutic intervention. Therapeutically, targeting TLRs is a promising strategy for SLE treatment, with drugs like hydroxychloroquine already in clinical use.
Collapse
Affiliation(s)
- Susannah von Hofsten
- Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway;
| | - Kristin Andreassen Fenton
- Centre of Clinical Research and Education, University Hospital of North Norway, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway;
| | - Hege Lynum Pedersen
- Centre of Clinical Research and Education, University Hospital of North Norway, Department of Medical Biology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9019 Tromsø, Norway;
| |
Collapse
|
5
|
Tremain AC, Wallace RP, Lorentz KM, Thornley TB, Antane JT, Raczy MR, Reda JW, Alpar AT, Slezak AJ, Watkins EA, Maulloo CD, Budina E, Solanki A, Nguyen M, Bischoff DJ, Harrington JL, Mishra R, Conley GP, Marlin R, Dereuddre-Bosquet N, Gallouët AS, LeGrand R, Wilson DS, Kontos S, Hubbell JA. Synthetically glycosylated antigens for the antigen-specific suppression of established immune responses. Nat Biomed Eng 2023; 7:1142-1155. [PMID: 37679570 DOI: 10.1038/s41551-023-01086-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 08/02/2023] [Indexed: 09/09/2023]
Abstract
Inducing antigen-specific tolerance during an established immune response typically requires non-specific immunosuppressive signalling molecules. Hence, standard treatments for autoimmunity trigger global immunosuppression. Here we show that established antigen-specific responses in effector T cells and memory T cells can be suppressed by a polymer glycosylated with N-acetylgalactosamine (pGal) and conjugated to the antigen via a self-immolative linker that allows for the dissociation of the antigen on endocytosis and its presentation in the immunoregulatory environment. We show that pGal-antigen therapy induces antigen-specific tolerance in a mouse model of experimental autoimmune encephalomyelitis (with programmed cell-death-1 and the co-inhibitory ligand CD276 driving the tolerogenic responses), as well as the suppression of antigen-specific responses to vaccination against a DNA-based simian immunodeficiency virus in non-human primates. Our findings show that pGal-antigen therapy invokes mechanisms of immune tolerance to resolve antigen-specific inflammatory T-cell responses and suggest that the therapy may be applicable across autoimmune diseases.
Collapse
Affiliation(s)
- Andrew C Tremain
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Rachel P Wallace
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | | | | | - Jennifer T Antane
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Michal R Raczy
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Joseph W Reda
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Aaron T Alpar
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Anna J Slezak
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Elyse A Watkins
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Chitavi D Maulloo
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Erica Budina
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Ani Solanki
- Animal Resources Center, University of Chicago, Chicago, IL, USA
| | - Mindy Nguyen
- Animal Resources Center, University of Chicago, Chicago, IL, USA
| | | | | | | | | | - Romain Marlin
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Nathalie Dereuddre-Bosquet
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Anne-Sophie Gallouët
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Roger LeGrand
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - D Scott Wilson
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Biomedical Engineering Department, Johns Hopkins University, Baltimore, MD, USA.
| | | | - Jeffrey A Hubbell
- Committee on Immunology, University of Chicago, Chicago, IL, USA.
- Pritzker School for Molecular Engineering, University of Chicago, Chicago, IL, USA.
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
6
|
Chen SN, Mai ZY, Mai JN, Liang W, Dong ZX, Ju FE, Chan SH, Fang Z, Xu Y, Uziel O, He C, Zhang XD, Zheng Y. E2F2 modulates cell adhesion through the transcriptional regulation of PECAM1 in multiple myeloma. Br J Haematol 2023; 202:840-855. [PMID: 37365680 DOI: 10.1111/bjh.18958] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/26/2023] [Accepted: 06/17/2023] [Indexed: 06/28/2023]
Abstract
Multiple myeloma (MM) is the second most common haematological malignancy. Despite the development of new drugs and treatments in recent years, the therapeutic outcomes of patients are not satisfactory. It is necessary to further investigate the molecular mechanism underlying MM progression. Herein, we found that high E2F2 expression was correlated with poor overall survival and advanced clinical stages in MM patients. Gain- and loss-of-function studies showed that E2F2 inhibited cell adhesion and consequently activated cell epithelial-to-mesenchymal transition (EMT) and migration. Further experiments revealed that E2F2 interacted with the PECAM1 promoter to suppress its transcriptional activity. The E2F2-knockdown-mediated promotion of cell adhesion was significantly reversed by the repression of PECAM1 expression. Finally, we observed that silencing E2F2 significantly inhibited viability and tumour progression in MM cell models and xenograft mouse models respectively. This study demonstrates that E2F2 plays a vital role as a tumour accelerator by inhibiting PECAM1-dependent cell adhesion and accelerating MM cell proliferation. Therefore, E2F2 may serve as a potential independent prognostic marker and therapeutic target for MM.
Collapse
Affiliation(s)
- Shu-Na Chen
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Zhi-Ying Mai
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Jun-Na Mai
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Weiyao Liang
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Zhao-Xia Dong
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Fei-Er Ju
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Sze-Hoi Chan
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Zhigang Fang
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yichuan Xu
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Orit Uziel
- The Felsenstein Medical Research Center, Institute of Hematology Rabin Medical Center and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Xing-Ding Zhang
- Key Laboratory for Efficacy and Safety Evaluation of Hematological Malignancy Targeted Medicine of Guangdong Provincial Drug Administration, School of Medicine, Sun Yat-Sen University, Shenzhen, China
| | - Yongjiang Zheng
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
7
|
Cui S, Shin YJ, Fang X, Lee H, Eum SH, Ko EJ, Lim SW, Shin E, Lee KI, Lee JY, Lee CB, Bae SK, Yang CW, Chung BH. CRISPR/Cas9-mediated A4GALT suppression rescues Fabry disease phenotypes in a kidney organoid model. Transl Res 2023:S1931-5244(23)00025-7. [PMID: 36805562 DOI: 10.1016/j.trsl.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 01/28/2023] [Accepted: 02/13/2023] [Indexed: 02/20/2023]
Abstract
The objective of this study was to investigate whether CRISPR/Cas9-mediated suppression of A4GALT could rescue phenotype of Fabry disease nephropathy (FDN) using human induced pluripotent stem cells (hiPSCs) derived kidney organoid system. We generated FDN patient-derived hiPSC (CMC-Fb-002) and FD-specific hiPSCs (GLA-KO) by knock-out (KO) of GLA in wild-type (WT) hiPSCs using CRISPR/Cas9. We then performed A4GALT KO in both CMC-Fb-002 and GLA-KO to make Fb-002-A4GALT-KO and GLA/A4GALT-KO, respectively. Using these hiPSCs, we generated kidney organoids and compared alpha-galactosidase-A enzyme (α-GalA) activity, globotriaosylceramide (Gb-3) deposition, and zebra body formation under electron microscopy (EM). We also compared mRNA expression levels using RNA-seq and qPCR. Generated hiPSCs showed typical pluripotency markers without chromosomal disruption. Expression levels of GLA in CMC-Fb-002 and GLA-KO and expression levels of A4GALT in Fb-002-A4GALT-KO and GLA/A4GALT-KO were successfully decreased compared to those in WT-hiPSCs, respectively. Generated kidney organoids using these hiPSCs expressed typical nephron markers. In CMC-Fb-002 and GLA-KO organoids, α-GalA activity was significantly decreased along with increased deposition of Gb-3 in comparison with WT organoids. Intralysosomal inclusion body was also detected under EM. However, these disease phenotypes were rescued by KO of A4GALT in both GLA/A4GALT-KO and Fb-002-A4GALT-KO kidney organoids. RNA-seq showed increased expression levels of genes related to FDN progression in both GLA-mutant organoids compared to those in WT. Such increases were rescued in GLA/A4GALT-KO or Fb-002-A4GALT-KO organoids. CRISPR/Cas9 mediated suppression of A4GALT could rescue FDN phenotype. Hence, it can be proposed as a therapeutic approach to treat FDN.
Collapse
Affiliation(s)
- Sheng Cui
- Transplantation Research Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoo Jin Shin
- Transplantation Research Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Xianying Fang
- Transplantation Research Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hanbi Lee
- Transplantation Research Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang Hun Eum
- Transplantation Research Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Division of Nephrology, Department of Internal Medicine, Incheon St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Jeong Ko
- Transplantation Research Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sun Woo Lim
- Transplantation Research Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | | | | | - Chae Bin Lee
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Soo Kyung Bae
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Chul Woo Yang
- Transplantation Research Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Byung Ha Chung
- Transplantation Research Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, The College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Ma S, Wang C, Khan A, Liu L, Dalgleish J, Kiryluk K, He Z, Ionita-Laza I. BIGKnock: fine-mapping gene-based associations via knockoff analysis of biobank-scale data. Genome Biol 2023; 24:24. [PMID: 36782330 PMCID: PMC9926792 DOI: 10.1186/s13059-023-02864-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 01/23/2023] [Indexed: 02/15/2023] Open
Abstract
We propose BIGKnock (BIobank-scale Gene-based association test via Knockoffs), a computationally efficient gene-based testing approach for biobank-scale data, that leverages long-range chromatin interaction data, and performs conditional genome-wide testing via knockoffs. BIGKnock can prioritize causal genes over proxy associations at a locus. We apply BIGKnock to the UK Biobank data with 405,296 participants for multiple binary and quantitative traits, and show that relative to conventional gene-based tests, BIGKnock produces smaller sets of significant genes that contain the causal gene(s) with high probability. We further illustrate its ability to pinpoint potential causal genes at [Formula: see text] of the associated loci.
Collapse
Affiliation(s)
- Shiyang Ma
- Department of Biostatistics, Columbia University, New York, NY, USA
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Wang
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Linxi Liu
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - James Dalgleish
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Zihuai He
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
9
|
Harley ITW, Allison K, Scofield RH. Polygenic autoimmune disease risk alleles impacting B cell tolerance act in concert across shared molecular networks in mouse and in humans. Front Immunol 2022; 13:953439. [PMID: 36090990 PMCID: PMC9450536 DOI: 10.3389/fimmu.2022.953439] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Most B cells produced in the bone marrow have some level of autoreactivity. Despite efforts of central tolerance to eliminate these cells, many escape to periphery, where in healthy individuals, they are rendered functionally non-responsive to restimulation through their antigen receptor via a process termed anergy. Broad repertoire autoreactivity may reflect the chances of generating autoreactivity by stochastic use of germline immunoglobulin gene segments or active mechanisms may select autoreactive cells during egress to the naïve peripheral B cell pool. Likewise, it is unclear why in some individuals autoreactive B cell clones become activated and drive pathophysiologic changes in autoimmune diseases. Both of these remain central questions in the study of the immune system(s). In most individuals, autoimmune diseases arise from complex interplay of genetic risk factors and environmental influences. Advances in genome sequencing and increased statistical power from large autoimmune disease cohorts has led to identification of more than 200 autoimmune disease risk loci. It has been observed that autoantibodies are detectable in the serum years to decades prior to the diagnosis of autoimmune disease. Thus, current models hold that genetic defects in the pathways that control autoreactive B cell tolerance set genetic liability thresholds across multiple autoimmune diseases. Despite the fact these seminal concepts were developed in animal (especially murine) models of autoimmune disease, some perceive a disconnect between human risk alleles and those identified in murine models of autoimmune disease. Here, we synthesize the current state of the art in our understanding of human risk alleles in two prototypical autoimmune diseases - systemic lupus erythematosus (SLE) and type 1 diabetes (T1D) along with spontaneous murine disease models. We compare these risk networks to those reported in murine models of these diseases, focusing on pathways relevant to anergy and central tolerance. We highlight some differences between murine and human environmental and genetic factors that may impact autoimmune disease development and expression and may, in turn, explain some of this discrepancy. Finally, we show that there is substantial overlap between the molecular networks that define these disease states across species. Our synthesis and analysis of the current state of the field are consistent with the idea that the same molecular networks are perturbed in murine and human autoimmune disease. Based on these analyses, we anticipate that murine autoimmune disease models will continue to yield novel insights into how best to diagnose, prognose, prevent and treat human autoimmune diseases.
Collapse
Affiliation(s)
- Isaac T. W. Harley
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
- Rheumatology Section, Medicine Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| | - Kristen Allison
- Division of Rheumatology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Human Immunology and Immunotherapy Initiative (HI3), Department of Immunology, University of Colorado School of Medicine, Aurora, CO, United States
| | - R. Hal Scofield
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Medical/Research Service, US Department of Veterans Affairs Medical Center, Oklahoma City, OK, United States
| |
Collapse
|
10
|
Mustafa N, Mitxelena J, Infante A, Zenarruzabeitia O, Eriz A, Iglesias-Ara A, Zubiaga AM. E2f2 Attenuates Apoptosis of Activated T Lymphocytes and Protects from Immune-Mediated Injury through Repression of Fas and FasL. Int J Mol Sci 2021; 23:ijms23010311. [PMID: 35008734 PMCID: PMC8745065 DOI: 10.3390/ijms23010311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/20/2021] [Accepted: 12/25/2021] [Indexed: 12/03/2022] Open
Abstract
Targeted disruption of E2f2 in mice causes T-cell hyperactivation and a disproportionate cell cycle entry upon stimulation. However, E2f2−/− mice do not develop a lymphoproliferative condition. We report that E2f2 plays a Fas-dependent anti-apoptotic function in vitro and in vivo. TCR-stimulated murine E2f2−/− T cells overexpress the proapoptotic genes Fas and FasL and exhibit enhanced apoptosis, which is prevented by treatment with neutralizing anti-FasL antibodies. p53 pathway is activated in TCR-stimulated E2f2−/− lymphocytes, but targeted disruption of p53 in E2f2−/− mice does not abrogate Fas/FasL expression or apoptosis, implying a p53-independent apoptotic mechanism. We show that E2f2 is recruited to Fas and FasL gene promoters to repress their expression. in vivo, E2f2−/− mice are prone to develop immune-mediated liver injury owing to an aberrant lymphoid Fas/FasL activation. Taken together, our results suggest that E2f2-dependent inhibition of Fas/FasL pathway may play a direct role in limiting the development of immune-mediated pathologies.
Collapse
Affiliation(s)
- Noor Mustafa
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48080 Bilbao, Spain; (N.M.); (J.M.); (A.E.)
| | - Jone Mitxelena
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48080 Bilbao, Spain; (N.M.); (J.M.); (A.E.)
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Arantza Infante
- Stem Cells and Cell Therapy Laboratory, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
| | - Ainhoa Eriz
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48080 Bilbao, Spain; (N.M.); (J.M.); (A.E.)
| | - Ainhoa Iglesias-Ara
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48080 Bilbao, Spain; (N.M.); (J.M.); (A.E.)
- Correspondence: (A.I.-A.); (A.M.Z.); Tel.: +34-94-601-5799 (A.I.-A.); +34-94-601-2603 (A.M.Z.); Fax: +34-94-601-3143 (A.M.Z.)
| | - Ana M. Zubiaga
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, 48080 Bilbao, Spain; (N.M.); (J.M.); (A.E.)
- Correspondence: (A.I.-A.); (A.M.Z.); Tel.: +34-94-601-5799 (A.I.-A.); +34-94-601-2603 (A.M.Z.); Fax: +34-94-601-3143 (A.M.Z.)
| |
Collapse
|
11
|
Lewis DA, Ly T. Cell Cycle Entry Control in Naïve and Memory CD8 + T Cells. Front Cell Dev Biol 2021; 9:727441. [PMID: 34692683 PMCID: PMC8526999 DOI: 10.3389/fcell.2021.727441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/07/2021] [Indexed: 12/20/2022] Open
Abstract
CD8+ T cells play important roles in immunity and immuno-oncology. Upon antigen recognition and co-stimulation, naïve CD8+ T cells escape from dormancy to engage in a complex programme of cellular growth, cell cycle entry and differentiation, resulting in rapid proliferation cycles that has the net effect of producing clonally expanded, antigen-specific cytotoxic T lymphocytes (CTLs). A fraction of activated T cells will re-enter dormancy by differentiating into memory T cells, which have essential roles in adaptive immunity. In this review, we discuss the current understanding of cell cycle entry control in CD8+ T cells and crosstalk between these mechanisms and pathways regulating immunological phenotypes.
Collapse
Affiliation(s)
- David A. Lewis
- Ashworth Laboratories, Institute of Immunology and Infectious Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Tony Ly
- Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Gene Regulation and Expression, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
12
|
Chakhtoura M, Fang M, Cubas R, O’Connor MH, Nichols CN, Richardson B, Talla A, Moir S, Cameron MJ, Tardif V, Haddad EK. Germinal Center T follicular helper (GC-Tfh) cell impairment in chronic HIV infection involves c-Maf signaling. PLoS Pathog 2021; 17:e1009732. [PMID: 34280251 PMCID: PMC8289045 DOI: 10.1371/journal.ppat.1009732] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
We have recently demonstrated that the function of T follicular helper (Tfh) cells from lymph nodes (LN) of HIV-infected individuals is impaired. We found that these cells were unable to provide proper help to germinal center (GC)-B cells, as observed by altered and inefficient anti-HIV antibody response and premature death of memory B cells. The underlying molecular mechanisms of this dysfunction remain poorly defined. Herein, we have used a unique transcriptional approach to identify these molecular defects. We consequently determined the transcriptional profiles of LN GC-Tfh cells following their interactions with LN GC-B cells from HIV-infected and HIV-uninfected individuals, rather than analyzing resting ex-vivo GC-Tfh cells. We observed that proliferating GC-Tfh cells from HIV-infected subjects were transcriptionally different than their HIV-uninfected counterparts, and displayed a significant downregulation of immune- and GC-Tfh-associated pathways and genes. Our results strongly demonstrated that MAF (coding for the transcription factor c-Maf) and its upstream signaling pathway mediators (IL6R and STAT3) were significantly downregulated in HIV-infected subjects, which could contribute to the impaired GC-Tfh and GC-B cell functions reported during infection. We further showed that c-Maf function was associated with the adenosine pathway and that the signaling upstream c-Maf could be partially restored by adenosine deaminase -1 (ADA-1) supplementation. Overall, we identified a novel mechanism that contributes to GC-Tfh cell impairment during HIV infection. Understanding how GC-Tfh cell function is altered in HIV is crucial and could provide critical information about the mechanisms leading to the development and maintenance of effective anti-HIV antibodies. Human immunodeficiency virus (HIV) remains a worldwide burden despite available treatments. The virus induces dysregulations in major immune cells and organs including lymph nodes. Germinal center T follicular helper (GC-Tfh) cells are immune cells which induce specific anti-HIV antibodies by helping GC-B cells. In chronic HIV, the interaction between these two cell types is defective, leading to modified and inefficient anti-HIV antibody responses. In this study, we examined the underlying mechanisms of this dysfunction. We observed that proliferating GC-Tfh cells from HIV-infected individuals, displayed distinctive gene expression than those from -uninfected subjects, following GC-B cell interaction. Furthermore, GC-Tfh cells from HIV patients showed a reduction in important immune-related pathway and gene expression. A number of essential GC-Tfh cell genes, such as MAF and its associated genes (IL6R and STAT3), were particularly attenuated in HIV, contributing to the impaired cells function. Moreover, we found an association between MAF function and the key enzyme adenosine deaminase-1 (ADA-1), where supplementation with ADA-1 partially restored the dysfunctional signaling in GC-Tfh cells during chronic infection. Understanding how GC-Tfh cells are altered in HIV is critical to elucidate the mechanisms leading to effective anti-HIV antibodies.
Collapse
Affiliation(s)
- Marita Chakhtoura
- Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Mike Fang
- Department of Population and Quantitative Health Services, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Rafael Cubas
- Iovance Biotherapeutics, San Carlos, California, United States of America
| | - Margaret H. O’Connor
- Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Molecular and Cellular Biology and Genetics, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Carmen N. Nichols
- Department of Population and Quantitative Health Services, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Brian Richardson
- Department of Population and Quantitative Health Services, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Aarthi Talla
- Allen Institute for Immunology, Seattle, Washington, United States of America
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mark J. Cameron
- Department of Population and Quantitative Health Services, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Virginie Tardif
- Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- Sorbonne University, INSERM, Center of Reasearch in Myology (Association Institut de Myologie) UMRS 974, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, Paris, France
- * E-mail: (VT); (EKH)
| | - Elias K. Haddad
- Department of Medicine, Division of Infectious Diseases & HIV Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (VT); (EKH)
| |
Collapse
|
13
|
Abstract
E2F transcription factor 2 (E2F2) is a member of the E2F family of transcription factors. The classical view is that some E2Fs act as "activators" and others "inhibitors" of cell cycle gene expression. However, the so-called "activator" E2F2 is particularly enigmatic, with seemingly contradictory roles in the cell cycle, proliferation, apoptosis, inflammation, and cell migration and invasion. How can we rationalize the apparently opposing functions of E2F2 in different situations? This is difficult because different methods of studying E2F2 have yielded conflicting results, so extrapolating mechanisms from an observed endpoint is challenging. This review will attempt to summarize and clarify these issues. This review focuses on genetic studies that have helped elucidate the biological functions of E2F2 and that have enhanced our understanding of how E2F2 is integrated into pathways controlling the cell cycle, proliferation, apoptosis, inflammation, and cell migration and invasion. This review will also discuss the function of E2F2 in cancer and other diseases. This review provides a strong basis for further research on the biological function and clinical potential of E2F2.
Collapse
Affiliation(s)
- Luwen Li
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University, Ji'nan, China.,Key Lab for Biotech-Drugs of National Health Commission, Ji'nan, China.,Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University, Ji'nan, China
| | - Shiguan Wang
- Medical College, Shandong University, Ji'nan, China
| | - Yihang Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University, Ji'nan, China.,Key Lab for Biotech-Drugs of National Health Commission, Ji'nan, China.,Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University, Ji'nan, China
| | - Jihong Pan
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University, Ji'nan, China.,Key Lab for Biotech-Drugs of National Health Commission, Ji'nan, China.,Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University, Ji'nan, China
| |
Collapse
|
14
|
Wang Y, Franks JM, Yang M, Toledo DM, Wood TA, Hinchcliff M, Whitfield ML. Regulator combinations identify systemic sclerosis patients with more severe disease. JCI Insight 2020; 5:137567. [PMID: 32721949 PMCID: PMC7526449 DOI: 10.1172/jci.insight.137567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/22/2020] [Indexed: 11/17/2022] Open
Abstract
Systemic sclerosis (SSc) is a heterogeneous autoimmune disorder that results in skin fibrosis, autoantibody production, and internal organ dysfunction. We previously identified 4 “intrinsic” subsets of SSc based upon skin gene expression that are found across organ systems. Gene expression regulators that underlie the SSc-intrinsic subsets, or are associated with clinical covariates, have not been systematically characterized. Here, we present a computational framework to calculate the activity scores of gene expression regulators and identify their associations with SSc clinical outcomes. We found that regulator activity scores can reproduce the intrinsic molecular subsets, with distinct sets of regulators identified for inflammatory, fibroproliferative, limited, and normal-like samples. Regulators most highly correlated with modified Rodnan skin score (MRSS) also varied by intrinsic subset. We identified subgroups of patients with fibroproliferative and inflammatory SSc with more severe pathophenotypes, such as higher MRSS and increased likelihood of interstitial lung disease (ILD). Using an independent cohort, we show that the group with more severe ILD was more likely to show forced vital capacity decline over a period of 36–54 months. Our results demonstrate an association among the activation of regulators, gene expression subsets, and clinical variables that can identify patients with SSc with more severe disease. An association between the activation of regulators, gene expression subsets, and clinical variables identifies systemic sclerosis patients with more severe disease.
Collapse
Affiliation(s)
- Yue Wang
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Jennifer M Franks
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Monica Yang
- Department of Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Diana M Toledo
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Tammara A Wood
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Monique Hinchcliff
- Department of Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Yale School of Medicine, Section of Allergy, Rheumatology and Immunology, New Haven, Connecticut, USA
| | - Michael L Whitfield
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| |
Collapse
|
15
|
Palmer N, Talib SZA, Kaldis P. Diverse roles for CDK-associated activity during spermatogenesis. FEBS Lett 2019; 593:2925-2949. [PMID: 31566717 PMCID: PMC6900092 DOI: 10.1002/1873-3468.13627] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/20/2019] [Accepted: 09/26/2019] [Indexed: 12/22/2022]
Abstract
The primary function of cyclin-dependent kinases (CDKs) in complex with their activating cyclin partners is to promote mitotic division in somatic cells. This canonical cell cycle-associated activity is also crucial for fertility as it allows the proliferation and differentiation of stem cells within the reproductive organs to generate meiotically competent cells. Intriguingly, several CDKs exhibit meiosis-specific functions and are essential for the completion of the two reductional meiotic divisions required to generate haploid gametes. These meiosis-specific functions are mediated by both known CDK/cyclin complexes and meiosis-specific CDK-regulators and are important for a variety of processes during meiotic prophase. The majority of meiotic defects observed upon deletion of these proteins occur during the extended prophase I of the first meiotic division. Importantly a lack of redundancy is seen within the meiotic arrest phenotypes described for many of these proteins, suggesting intricate layers of cell cycle control are required for normal meiotic progression. Using the process of male germ cell development (spermatogenesis) as a reference, this review seeks to highlight the diverse roles of selected CDKs their activators, and their regulators during gametogenesis.
Collapse
Affiliation(s)
- Nathan Palmer
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.,Department of Biochemistry, National University of Singapore (NUS), Singapore, Singapore
| | - S Zakiah A Talib
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.,Department of Biochemistry, National University of Singapore (NUS), Singapore, Singapore.,Department of Clinical Sciences, Clinical Research Centre (CRC), Lund University, Malmö, Sweden
| |
Collapse
|
16
|
Hollern DP, Swiatnicki MR, Rennhack JP, Misek SA, Matson BC, McAuliff A, Gallo KA, Caron KM, Andrechek ER. E2F1 Drives Breast Cancer Metastasis by Regulating the Target Gene FGF13 and Altering Cell Migration. Sci Rep 2019; 9:10718. [PMID: 31341204 PMCID: PMC6656723 DOI: 10.1038/s41598-019-47218-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 07/12/2019] [Indexed: 12/27/2022] Open
Abstract
In prior work we demonstrated that loss of E2F transcription factors inhibits metastasis. Here we address the mechanisms for this phenotype and identify the E2F regulated genes that coordinate tumor cell metastasis. Transcriptomic profiling of E2F1 knockout tumors identified a role for E2F1 as a master regulator of a suite of pro-metastatic genes, but also uncovered E2F1 target genes with an unknown role in pulmonary metastasis. High expression of one of these genes, Fgf13, is associated with early human breast cancer metastasis in a clinical dataset. Together these data led to the hypothesis that Fgf13 is critical for breast cancer metastasis, and that upregulation of Fgf13 may partially explain how E2F1 promotes breast cancer metastasis. To test this hypothesis we ablated Fgf13 via CRISPR. Deletion of Fgf13 in a MMTV-PyMT breast cancer cell line reduces colonization of the lungs in a tail vein injection. In addition, loss of Fgf13 reduced in vitro cell migration, suggesting that Fgf13 may be critical for tumor cells to escape the primary tumor and to colonize the distal sites. The significance of this work is twofold: we have both uncovered genomic features by which E2F1 regulates metastasis and we have identified new pro-metastatic functions for the E2F1 target gene Fgf13.
Collapse
Affiliation(s)
- Daniel P Hollern
- Lineberger Comprehensive Cancer Center University of North Carolina, Chapel Hill, United States
| | - Matthew R Swiatnicki
- Department of Physiology, Michigan State University, East Lansing, United States
| | - Jonathan P Rennhack
- Department of Physiology, Michigan State University, East Lansing, United States
| | - Sean A Misek
- Department of Physiology, Michigan State University, East Lansing, United States
| | - Brooke C Matson
- University of North Carolina Department of Cell Biology, Chapel Hill, United States
| | - Andrew McAuliff
- Department of Physiology, Michigan State University, East Lansing, United States
| | - Kathleen A Gallo
- Department of Physiology, Michigan State University, East Lansing, United States
| | - Kathleen M Caron
- Lineberger Comprehensive Cancer Center University of North Carolina, Chapel Hill, United States
| | - Eran R Andrechek
- Department of Physiology, Michigan State University, East Lansing, United States.
| |
Collapse
|
17
|
Caudell DL, Michalson KT, Andrews RN, Snow WW, Bourland JD, DeBo RJ, Cline JM, Sempowski GD, Register TC. Transcriptional Profiling of Non-Human Primate Lymphoid Organ Responses to Total-Body Irradiation. Radiat Res 2019; 192:40-52. [PMID: 31059377 PMCID: PMC6699496 DOI: 10.1667/rr15100.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The global threat of exposure to radiation and its subsequent outcomes require the development of effective strategies to mitigate immune cell injury. In this study we explored transcriptional and immunophenotypic characteristics of lymphoid organs of a non-human primate model after total-body irradiation (TBI). Fifteen middle-aged adult, ovariectomized, female cynomolgus macaques received a single dose of 0, 2 or 5 Gy gamma radiation. Thymus, spleen and lymph node from three controls and 2 Gy (n = 2) and 5 Gy (n = 2) exposed animals were assessed for molecular responses to TBI through microarray-based transcriptional profiling at day 5 postirradiation, and cellular changes through immunohistochemical (IHC) characterization of markers for B and T lymphocytes and macrophages across all 15 animals at time points up to 6 months postirradiation. Irradiated macaques developed acute hematopoietic syndrome. Analysis of array data at day 5 postirradiation identified transcripts with ≥2-fold difference from control and a false discovery rate (FDR) of Padj < 0.05 in lymph node (n = 666), spleen (n = 493) and thymus (n=3,014). Increasing stringency of the FDR to P < 0.001 reduced the number of genes to 71 for spleen and 379 for thymus. IHC and gene expression data demonstrated that irradiated animals had reduced numbers of T and B lymphocytes along with relative elevations of macrophages. Transcriptional analysis revealed unique patterns in primary and secondary lymphoid organs of cynomolgus macaques. Among the many differentially regulated transcripts, upregulation of noncoding RNAs [MIR34A for spleen and thymus and NEAT1 (NCRNA00084) for thymus] showed potential as biomarkers of radiation injury and targets for mitigating the effects of radiation-induced hematopoietic syndrome-impaired lymphoid reconstitution.
Collapse
Affiliation(s)
- David L. Caudell
- Departments of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Kristofer T. Michalson
- Departments of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Rachel N. Andrews
- Departments of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - William W. Snow
- Departments of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - J. Daniel Bourland
- Departments of Radiation Oncology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Ryne J. DeBo
- Departments of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - J. Mark Cline
- Departments of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Gregory D. Sempowski
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Thomas C. Register
- Departments of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| |
Collapse
|
18
|
Cuitiño MC, Pécot T, Sun D, Kladney R, Okano-Uchida T, Shinde N, Saeed R, Perez-Castro AJ, Webb A, Liu T, Bae SI, Clijsters L, Selner N, Coppola V, Timmers C, Ostrowski MC, Pagano M, Leone G. Two Distinct E2F Transcriptional Modules Drive Cell Cycles and Differentiation. Cell Rep 2019; 27:3547-3560.e5. [PMID: 31130414 PMCID: PMC6673649 DOI: 10.1016/j.celrep.2019.05.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/14/2019] [Accepted: 04/30/2019] [Indexed: 02/05/2023] Open
Abstract
Orchestrating cell-cycle-dependent mRNA oscillations is critical to cell proliferation in multicellular organisms. Even though our understanding of cell-cycle-regulated transcription has improved significantly over the last three decades, the mechanisms remain untested in vivo. Unbiased transcriptomic profiling of G0, G1-S, and S-G2-M sorted cells from FUCCI mouse embryos suggested a central role for E2Fs in the control of cell-cycle-dependent gene expression. The analysis of gene expression and E2F-tagged knockin mice with tissue imaging and deep-learning tools suggested that post-transcriptional mechanisms universally coordinate the nuclear accumulation of E2F activators (E2F3A) and canonical (E2F4) and atypical (E2F8) repressors during the cell cycle in vivo. In summary, we mapped the spatiotemporal expression of sentinel E2F activators and canonical and atypical repressors at the single-cell level in vivo and propose that two distinct E2F modules relay the control of gene expression in cells actively cycling (E2F3A-8-4) and exiting the cycle (E2F3A-4) during mammalian development.
Collapse
Affiliation(s)
- Maria C Cuitiño
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Thierry Pécot
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Daokun Sun
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Molecular Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Raleigh Kladney
- Department of Molecular Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Takayuki Okano-Uchida
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Neelam Shinde
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Resham Saeed
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Antonio J Perez-Castro
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Amy Webb
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Tom Liu
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Soo In Bae
- Department of Molecular Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Linda Clijsters
- Department of Biochemistry and Molecular Pharmacology, Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | - Nicholas Selner
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Vincenzo Coppola
- Department of Molecular Genetics, Ohio State University, Columbus, OH 43210, USA
| | - Cynthia Timmers
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Michael C Ostrowski
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Gustavo Leone
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
19
|
Kim HR, Rahman FU, Kim KS, Kim EK, Cho SM, Lee K, Moon OS, Seo YW, Yoon WK, Won YS, Kang H, Kim HC, Nam KH. Critical Roles of E2F3 in Growth and Musculo-skeletal Phenotype in Mice. Int J Med Sci 2019; 16:1557-1563. [PMID: 31839743 PMCID: PMC6909802 DOI: 10.7150/ijms.39068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/11/2019] [Indexed: 12/24/2022] Open
Abstract
E2F3, a member of the E2F family, plays a critical role in cell cycle and proliferation by targeting downstream, retinoblastoma (RB) a tumor suppressor family protein. The purpose of this study, was to investigate the role and function of E2F3 in vivo. We examined phenotypic abnormalities, by deletion of the E2f3 gene in mice. Complete ablation of the E2F3 was fully penetrant, in the pure C57BL/6N background. The E2f3+/ - mouse embryo developed normally without fatal disorder. However, they exhibited reduced body weight, growth retardation, skeletal imperfection, and poor grip strength ability. Findings suggest that E2F3 has a pivotal role in muscle and bone development, and affect normal mouse growth.
Collapse
Affiliation(s)
- Hae-Rim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| | - Faiz Ur Rahman
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| | - Kwang-Soo Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea.,Department of Animal Science and Technology, Chung-Ang University, Seodong-daero 4726, Gyeonggi 17546, Korea
| | - Eun-Kyeung Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| | - Sang-Mi Cho
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| | - Kihoon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| | - Ok-Sung Moon
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| | - Young-Won Seo
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| | - Won-Kee Yoon
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| | - Young-Suk Won
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| | - Hoyoung Kang
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| | - Hyoung-Chin Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| | - Ki-Hoan Nam
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Yeonjudanji-ro 30, Chungbuk 28116, Korea
| |
Collapse
|
20
|
Nuzziello N, Vilardo L, Pelucchi P, Consiglio A, Liuni S, Trojano M, Liguori M. Investigating the Role of MicroRNA and Transcription Factor Co-regulatory Networks in Multiple Sclerosis Pathogenesis. Int J Mol Sci 2018; 19:ijms19113652. [PMID: 30463275 PMCID: PMC6274935 DOI: 10.3390/ijms19113652] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 01/04/2023] Open
Abstract
MicroRNAs (miRNAs) and transcription factors (TFs) play key roles in complex multifactorial diseases like multiple sclerosis (MS). Starting from the miRNomic profile previously associated with a cohort of pediatric MS (PedMS) patients, we applied a combined molecular and computational approach in order to verify published data in patients with adult-onset MS (AOMS). Six out of the 13 selected miRNAs (miR-320a, miR-125a-5p, miR-652-3p, miR-185-5p, miR-942-5p, miR-25-3p) were significantly upregulated in PedMS and AOMS patients, suggesting that they may be considered circulating biomarkers distinctive of the disease independently from age. A computational and unbiased miRNA-based screening of target genes not necessarily associated to MS was then performed in order to provide an extensive view of the genetic mechanisms underlying the disease. A comprehensive MS-specific miRNA-TF co-regulatory network was hypothesized; among others, SP1, RELA, NF-κB, TP53, AR, MYC, HDAC1, and STAT3 regulated the transcription of 61 targets. Interestingly, NF-κB and STAT3 cooperatively regulate the expression of immune response genes and control the cross-talk between inflammatory and immune cells. Further functional analysis will be performed on the identified critical hubs. Above all, in our view, this approach supports the need of multidisciplinary strategies for shedding light into the pathogenesis of MS.
Collapse
Affiliation(s)
- Nicoletta Nuzziello
- National Research Council, Institute of Biomedical Technologies, Bari Unit, 70126 Bari, Italy.
| | - Laura Vilardo
- National Research Council, Institute of Biomedical Technologies, Segrate Unit, 20090 Milan, Italy.
| | - Paride Pelucchi
- National Research Council, Institute of Biomedical Technologies, Segrate Unit, 20090 Milan, Italy.
| | - Arianna Consiglio
- National Research Council, Institute of Biomedical Technologies, Bari Unit, 70126 Bari, Italy.
| | - Sabino Liuni
- National Research Council, Institute of Biomedical Technologies, Bari Unit, 70126 Bari, Italy.
| | - Maria Trojano
- Department of Basic Sciences, Neurosciences and Sense Organs, University of Bari, 70124 Bari, Italy.
| | - Maria Liguori
- National Research Council, Institute of Biomedical Technologies, Bari Unit, 70126 Bari, Italy.
| |
Collapse
|
21
|
Wang S, Wang L, Wu C, Sun S, Pan JH. E2F2 directly regulates the STAT1 and PI3K/AKT/NF-κB pathways to exacerbate the inflammatory phenotype in rheumatoid arthritis synovial fibroblasts and mouse embryonic fibroblasts. Arthritis Res Ther 2018; 20:225. [PMID: 30286793 PMCID: PMC6235203 DOI: 10.1186/s13075-018-1713-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/04/2018] [Indexed: 11/30/2022] Open
Abstract
Background Expression of E2F transcription factor 2 (E2F2), a transcription factor related to the cell cycle, is abnormally high in rheumatoid arthritis synovial fibroblasts (RASFs). Deregulated expression of E2F2 leads to abnormal production of proinflammatory cytokines, such as interleukin (IL)-1α, IL-1β, and tumor necrosis factor (TNF)-α in RASFs. However, the underlying mechanism by which E2F2 regulates expression of IL-1α, IL-1β, and TNF-α has not been fully elucidated. This study aimed to elucidate this mechanism and confirm the pathological roles of E2F2 in rheumatoid arthritis (RA). Methods E2f2 knockout (KO) and wild-type (WT) mice were injected with collagen to induce RA. Cytokine production was assessed by quantitative real-time polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA). Western blot and qRT-PCR were performed to evaluate the effect of E2F2 on signaling pathway activity. Chromatin immunoprecipitation (ChIP)-PCR and luciferase assays were used to detect the transcriptional activity of target genes of E2F2. Nuclear translocation of STAT1 and p65 were assayed by Western blot, co-immunoprecipitation (co-IP), and immunofluorescence experiments. Results The occurrence and severity of collagen-induced arthritis were decreased in E2f2-KO mice compared with WT mice. The expression of IL-1α, IL-1β, and TNF-α was also suppressed in mouse embryonic fibroblasts (MEFs) from E2f2-KO mice and RASFs with E2F2 knocked down. Mechanistically, we found that E2F2 can upregulate the expression of STAT1 and MyD88 through direct binding to their promoters, facilitate the formation of STAT1/MyD88 complexes, and consequently activate AKT. However, silencing STAT1/MyD88 or inactivating AKT significantly attenuated the induction of IL-1α, IL-1β, and TNF-α caused by the introduction of E2F2. Conclusions This study confirms the pathological role of E2F2 in RA and found that the E2F2-STAT1/MyD88-Akt axis is closely related with the inflammatory phenotype in RASFs.
Collapse
Affiliation(s)
- Shiguan Wang
- Medical and Life Science College, University of Jinan, Jinan, 250062, Shandong, China.,Shandong Medicinal Biotechnology Centre, Jingshi Road, Jinan, 250000, Shandong, China.,Key Lab for Biotechnology Drugs of Ministry of Health, Jinan, 250000, Shandong, China
| | - Lin Wang
- Shandong Medicinal Biotechnology Centre, Jingshi Road, Jinan, 250000, Shandong, China.,Key Lab for Biotechnology Drugs of Ministry of Health, Jinan, 250000, Shandong, China.,Key Lab for Rare & Uncommon Diseases, Jinan, 250000, Shandong, China
| | - Changshun Wu
- Shandong Provincial Hospital affiliated to Shandong University, Jinan, 250000, Shandong, China
| | - Shui Sun
- Shandong Provincial Hospital affiliated to Shandong University, Jinan, 250000, Shandong, China
| | - Ji-Hong Pan
- Shandong Medicinal Biotechnology Centre, Jingshi Road, Jinan, 250000, Shandong, China. .,Key Lab for Biotechnology Drugs of Ministry of Health, Jinan, 250000, Shandong, China. .,Key Lab for Rare & Uncommon Diseases, Jinan, 250000, Shandong, China.
| |
Collapse
|
22
|
Increased expression of microRNAs, miR-20a and miR-326 in PBMCs of patients with type 1 diabetes. Mol Biol Rep 2018; 45:1973-1980. [PMID: 30194557 DOI: 10.1007/s11033-018-4352-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 08/31/2018] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disorder which is characterized by autoimmune attack on β cells of pancreas and lack of insulin. The involvement of microRNAs (miRNAs) in the development of immune system and their differential expression in various autoimmune diseases including T1D have been well established. In this study, the association between expression levels of miR-20a, miR-326 and T1D were evaluated. The expression levels of miR-20a and miR-326 were measured in the PBMCs of 21 T1D patients and 16 healthy controls using qPCR method. In silico analysis was also performed on targetome of miR-20a and miR-326. Both miR-20a (p value: 0.015) and miR-326 (p value: 0.005) were upregulated in the PBMCs of T1D patients compared to healthy controls. Furthermore, different dysregulated miR326-mRNA and miR20a-mRNA interactions were also suggested using integrative computational analysis. The expression level of miR-20a and miR-326 indicates significant association with T1D which suggests the possible regulatory effects of these non-coding RNAs in T1D.
Collapse
|
23
|
To B, Andrechek ER. Transcription factor compensation during mammary gland development in E2F knockout mice. PLoS One 2018; 13:e0194937. [PMID: 29617434 PMCID: PMC5884531 DOI: 10.1371/journal.pone.0194937] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 03/13/2018] [Indexed: 11/21/2022] Open
Abstract
The E2F transcription factors control key elements of development, including mammary gland branching morphogenesis, with several E2Fs playing essential roles. Additional prior data has demonstrated that loss of individual E2Fs can be compensated by other E2F family members, but this has not been tested in a mammary gland developmental context. Here we have explored the role of the E2Fs and their ability to functionally compensate for each other during mammary gland development. Using gene expression from terminal end buds and chromatin immunoprecipitation data for E2F1, E2F2 and E2F3, we noted both overlapping and unique mammary development genes regulated by each of the E2Fs. Based on our computational findings and the fact that E2Fs share a common binding motif, we hypothesized that E2F transcription factors would compensate for each other during mammary development and function. To test this hypothesis, we generated RNA from E2F1-/-, E2F2-/- and E2F3+/- mouse mammary glands. QRT-PCR on mammary glands during pregnancy demonstrated increases in E2F2 and E2F3a in the E2F1-/- mice and an increase in E2F2 levels in E2F3+/- mice. During lactation we noted that E2F3b transcript levels were increased in the E2F2-/- mice. Given that E2Fs have previously been noted to have the most striking effects on development during puberty, we hypothesized that loss of individual E2Fs would be compensated for at that time. Double mutant mice were generated and compared with the single knockouts. Loss of both E2F1 and E2F2 revealed a more striking phenotype than either knockout alone, indicating that E2F2 was compensating for E2F1 loss. Interestingly, while E2F2 was not able to functionally compensate for E2F3+/- during mammary outgrowth, increased E2F2 expression was observed in E2F3+/- mammary glands during pregnancy day 14.5 and lactation day 5. Together, these findings illustrate the specificity of E2F family members to compensate during development of the mammary gland.
Collapse
Affiliation(s)
- Briana To
- Department of Physiology, Michigan State University, East Lansing, MI, United States of America
| | - Eran R. Andrechek
- Department of Physiology, Michigan State University, East Lansing, MI, United States of America
- * E-mail:
| |
Collapse
|
24
|
Wei D, Li A, Zhao C, Wang H, Mei C, Khan R, Zan L. Transcriptional Regulation by CpG Sites Methylation in the Core Promoter Region of the Bovine SIX1 Gene: Roles of Histone H4 and E2F2. Int J Mol Sci 2018; 19:ijms19010213. [PMID: 29337851 PMCID: PMC5796162 DOI: 10.3390/ijms19010213] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/02/2018] [Accepted: 01/09/2018] [Indexed: 01/11/2023] Open
Abstract
DNA methylation is a major epigenetic modification of the genome and has an essential role in muscle development. The SIX1 gene is thought to play a principal role in mediating skeletal muscle development. In the present study, we determined that bovine SIX1 expression levels were significantly higher in the fetal bovine group (FB) and in undifferentiated Qinchuan cattle muscle cells (QCMCs) than in the adult bovine group (AB) and in differentiated QCMCs. Moreover, a bisulfite sequencing polymerase chain reaction (BSP) analysis of DNA methylation levels showed that three CpG sites in the core promoter region (−216/−28) of the bovine SIX1 gene exhibited significantly higher DNA methylation levels in the AB and differentiated QCMCs groups. In addition, we found that DNA methylation of SIX1 core promoter in vitro obviously influences the promoter activities. An electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assay, in combination with site-directed mutation and siRNA interference, demonstrated that histone H4 and E2F2 bind to the −216/−28 region and play important roles in SIX1 methylation regulation during development. The results of this study provide the foundation for a better understanding of the regulation of bovine SIX1 expression via methylation and muscle developmental in beef cattle.
Collapse
Affiliation(s)
- Dawei Wei
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Anning Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Chunping Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Hongbao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Chugang Mei
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Rajwali Khan
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
25
|
Seu L, Tidwell C, Timares L, Duverger A, Wagner FH, Goepfert PA, Westfall AO, Sabbaj S, Kutsch O. CD151 Expression Is Associated with a Hyperproliferative T Cell Phenotype. THE JOURNAL OF IMMUNOLOGY 2017; 199:3336-3347. [PMID: 28954890 DOI: 10.4049/jimmunol.1700648] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
Abstract
The tetraspanin CD151 is a marker of aggressive cell proliferation and invasiveness for a variety of cancer types. Given reports of CD151 expression on T cells, we explored whether CD151 would mark T cells in a hyperactivated state. Consistent with the idea that CD151 could mark a phenotypically distinct T cell subset, it was not uniformly expressed on T cells. CD151 expression frequency was a function of the T cell lineage (CD8 > CD4) and a function of the memory differentiation state (naive T cells < central memory T cells < effector memory T cells < T effector memory RA+ cells). CD151 and CD57, a senescence marker, defined the same CD28- T cell populations. However, CD151 also marked a substantial CD28+ T cell population that was not marked by CD57. Kinome array analysis demonstrated that CD28+CD151+ T cells form a subpopulation with a distinct molecular baseline and activation phenotype. Network analysis of these data revealed that cell cycle control and cell death were the most altered process motifs in CD28+CD151+ T cells. We demonstrate that CD151 in T cells is not a passive marker, but actively changed the cell cycle control and cell death process motifs of T cells. Consistent with these data, long-term T cell culture experiments in the presence of only IL-2 demonstrated that independent of their CD28 expression status, CD151+ T cells, but not CD151- T cells, would exhibit an Ag-independent, hyperresponsive proliferation phenotype. Not unlike its reported function as a tumor aggressiveness marker, CD151 in humans thus marks and enables hyperproliferative T cells.
Collapse
Affiliation(s)
- Lillian Seu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Christopher Tidwell
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Laura Timares
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Alexandra Duverger
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Frederic H Wagner
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Paul A Goepfert
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Andrew O Westfall
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Steffanie Sabbaj
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Olaf Kutsch
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
26
|
Petersen F, Yue X, Riemekasten G, Yu X. Dysregulated homeostasis of target tissues or autoantigens - A novel principle in autoimmunity. Autoimmun Rev 2017; 16:602-611. [PMID: 28411168 DOI: 10.1016/j.autrev.2017.04.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 03/11/2017] [Indexed: 01/22/2023]
Abstract
Monogenic autoimmune disorders provide a powerful tool for our understanding of the principles of autoimmunity due to the obvious impact of a single gene on the disease. So far, approximately 100 single gene defects causing murine monogenic autoimmune disorders have been reported and the functional characterization of these genes will provide significant progress in understanding the nature of autoimmunity. According to their function, genes leading to monogenic autoimmune disorders can be categorized into two groups. An expectable first group contains genes involved in the homeostasis of the immune system, including homeostasis of immune organs and immune cells. Intriguingly, the second group consists of genes functionally involved in the homeostasis of target tissues or autoantigens. According to our novel hypothesis, we propose that autoimmunity represents a consequence of a dysregulated homeostasis of the immune system and/or its targets including autoantigens and target tissues. In this review we refer to both aspects of homeostasis in autoimmunity with a highlight on the role of the homeostasis of target tissues and autoantigens.
Collapse
Affiliation(s)
- Frank Petersen
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Xiaoyang Yue
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Gabriela Riemekasten
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), 23845 Borstel, Germany; Department of Rheumatology, University of Lübeck, 23538 Lübeck, Germany
| | - Xinhua Yu
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), 23845 Borstel, Germany; Xiamen-Borstel Joint Laboratory of Autoimmunity, Medical College of Xiamen University, Xiamen 361102, China.
| |
Collapse
|
27
|
Hao Z, Sheng Y, Duncan GS, Li WY, Dominguez C, Sylvester J, Su YW, Lin GHY, Snow BE, Brenner D, You-Ten A, Haight J, Inoue S, Wakeham A, Elford A, Hamilton S, Liang Y, Zúñiga-Pflücker JC, He HH, Ohashi PS, Mak TW. K48-linked KLF4 ubiquitination by E3 ligase Mule controls T-cell proliferation and cell cycle progression. Nat Commun 2017; 8:14003. [PMID: 28084302 PMCID: PMC5241832 DOI: 10.1038/ncomms14003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 11/21/2016] [Indexed: 12/14/2022] Open
Abstract
T-cell proliferation is regulated by ubiquitination but the underlying molecular mechanism remains obscure. Here we report that Lys-48-linked ubiquitination of the transcription factor KLF4 mediated by the E3 ligase Mule promotes T-cell entry into S phase. Mule is elevated in T cells upon TCR engagement, and Mule deficiency in T cells blocks proliferation because KLF4 accumulates and drives upregulation of its transcriptional targets E2F2 and the cyclin-dependent kinase inhibitors p21 and p27. T-cell-specific Mule knockout (TMKO) mice develop exacerbated experimental autoimmune encephalomyelitis (EAE), show impaired generation of antigen-specific CD8+ T cells with reduced cytokine production, and fail to clear LCMV infections. Thus, Mule-mediated ubiquitination of the novel substrate KLF4 regulates T-cell proliferation, autoimmunity and antiviral immune responses in vivo. The E3 ligase Mule has been previously reported to be essential for B cell development and function by modulating p53 ubiquitination and degradation. Here Hao et al. identify KLF4 as a novel ubiquitination target of Mule and show it controls T cell proliferation and autoimmunity.
Collapse
Affiliation(s)
- Zhenyue Hao
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 2M9.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada M5G 2C1.,The Donnelly Centre for Cellular and Biomolecular Research, Banting and Best Department of Medical Research, and Department of Molecular Genetics, University of Toronto, 160 College Street, Toronto, Ontario, Canada M5S3E1
| | - Yi Sheng
- Department of Biology, York University, Toronto, Ontario, Canada M3J 1P3
| | - Gordon S Duncan
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Wanda Y Li
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Carmen Dominguez
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Jennifer Sylvester
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Yu-Wen Su
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9.,Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli County 35053, Taiwan
| | - Gloria H Y Lin
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Bryan E Snow
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Dirk Brenner
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, 29, rue Henri Koch, Esch-sur-Alzette L-4354, Luxembourg.,Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense DK-5000 Denmark
| | - Annick You-Ten
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Jillian Haight
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Satoshi Inoue
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Andrew Wakeham
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Alisha Elford
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Sara Hamilton
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Yi Liang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 2M9
| | - Juan C Zúñiga-Pflücker
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada M5G 2C1.,Sunnybrook and Women's College Health Sciences Centre, Toronto, Ontario, Canada M4N 3M5
| | - Housheng Hansen He
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 2M9.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada M5G 2C1
| | - Pamela S Ohashi
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 2M9.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada M5G 2C1.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada M5G 2C1
| | - Tak W Mak
- The Campbell Family Institute for Breast Cancer Research, University Health Network, Toronto, Ontario, Canada M5G 2M9.,Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada M5G 2M9.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada M5G 2C1.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada M5G 2C1
| |
Collapse
|
28
|
Safari-Alighiarloo N, Rezaei-Tavirani M, Taghizadeh M, Tabatabaei SM, Namaki S. Network-based analysis of differentially expressed genes in cerebrospinal fluid (CSF) and blood reveals new candidate genes for multiple sclerosis. PeerJ 2016; 4:e2775. [PMID: 28028462 PMCID: PMC5183126 DOI: 10.7717/peerj.2775] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/08/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The involvement of multiple genes and missing heritability, which are dominant in complex diseases such as multiple sclerosis (MS), entail using network biology to better elucidate their molecular basis and genetic factors. We therefore aimed to integrate interactome (protein-protein interaction (PPI)) and transcriptomes data to construct and analyze PPI networks for MS disease. METHODS Gene expression profiles in paired cerebrospinal fluid (CSF) and peripheral blood mononuclear cells (PBMCs) samples from MS patients, sampled in relapse or remission and controls, were analyzed. Differentially expressed genes which determined only in CSF (MS vs. control) and PBMCs (relapse vs. remission) separately integrated with PPI data to construct the Query-Query PPI (QQPPI) networks. The networks were further analyzed to investigate more central genes, functional modules and complexes involved in MS progression. RESULTS The networks were analyzed and high centrality genes were identified. Exploration of functional modules and complexes showed that the majority of high centrality genes incorporated in biological pathways driving MS pathogenesis. Proteasome and spliceosome were also noticeable in enriched pathways in PBMCs (relapse vs. remission) which were identified by both modularity and clique analyses. Finally, STK4, RB1, CDKN1A, CDK1, RAC1, EZH2, SDCBP genes in CSF (MS vs. control) and CDC37, MAP3K3, MYC genes in PBMCs (relapse vs. remission) were identified as potential candidate genes for MS, which were the more central genes involved in biological pathways. DISCUSSION This study showed that network-based analysis could explicate the complex interplay between biological processes underlying MS. Furthermore, an experimental validation of candidate genes can lead to identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Nahid Safari-Alighiarloo
- Proteomics Research Center, Department of Basic Science, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Department of Basic Science, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Mohammad Taghizadeh
- Bioinformatics Department, Institute of Biochemistry and Biophysics, Tehran University , Tehran , Iran
| | - Seyyed Mohammad Tabatabaei
- Medical Informatics Department, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Saeed Namaki
- Immunology Department, Faculty of Medical Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| |
Collapse
|
29
|
Increased metastasis with loss of E2F2 in Myc-driven tumors. Oncotarget 2016; 6:38210-24. [PMID: 26474282 PMCID: PMC4741994 DOI: 10.18632/oncotarget.5690] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/30/2015] [Indexed: 12/21/2022] Open
Abstract
In human breast cancer, mortality is associated with metastasis to distant sites. Therefore, it is critical to elucidate the biological mechanisms that underlie tumor progression and metastasis. Using signaling pathway signatures we previously predicted a role for E2F transcription factors in Myc induced tumors. To test this role we interbred MMTV-Myc transgenic mice with E2F knockouts. Surprisingly, we observed that the loss of E2F2 sharply increased the percentage of lung metastasis in MMTV-Myc transgenic mice. Examining the gene expression profile from these tumors, we identified genetic components that were potentially involved in mediating metastasis. These genes were filtered to uncover the genes involved in metastasis that also impacted distant metastasis free survival in human breast cancer. In order to elucidate the mechanism by which E2F2 loss enhanced metastasis we generated knockdowns of E2F2 in MDA-MB-231 cells and observed increased migration in vitro and increased lung colonization in vivo. We then examined genes that were differentially regulated between tumors from MMTV-Myc, MMTV-Myc E2F2−/−, and lung metastases samples and identified PTPRD. To test the role of PTPRD in E2F2-mediated breast cancer metastasis, we generated a knockdown of PTPRD in MDA-MB-231 cells. We noted that decreased levels of PTPRD resulted in decreased migration in vitro and decreased lung colonization in vivo. Taken together, these data indicate that E2F2 loss results in increased metastasis in breast cancer, potentially functioning through a PTPRD dependent mechanism.
Collapse
|
30
|
Zhang J, Loyd MR, Randall MS, Morris JJ, Shah JG, Ney PA. Repression by RB1 characterizes genes involved in the penultimate stage of erythroid development. Cell Cycle 2016; 14:3441-53. [PMID: 26397180 DOI: 10.1080/15384101.2015.1090067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Retinoblastoma-1 (RB1), and the RB1-related proteins p107 and p130, are key regulators of the cell cycle. Although RB1 is required for normal erythroid development in vitro, it is largely dispensable for erythropoiesis in vivo. The modest phenotype caused by RB1 deficiency in mice raises questions about redundancy within the RB1 family, and the role of RB1 in erythroid differentiation. Here we show that RB1 is the major pocket protein that regulates terminal erythroid differentiation. Erythroid cells lacking all pocket proteins exhibit the same cell cycle defects as those deficient for RB1 alone. RB1 has broad repressive effects on gene transcription in erythroid cells. As a group, RB1-repressed genes are generally well expressed but downregulated at the final stage of erythroid development. Repression correlates with E2F binding, implicating E2Fs in the recruitment of RB1 to repressed genes. Merging differential and time-dependent changes in expression, we define a group of approximately 800 RB1-repressed genes. Bioinformatics analysis shows that this list is enriched for terms related to the cell cycle, but also for terms related to terminal differentiation. Some of these have not been previously linked to RB1. These results expand the range of processes potentially regulated by RB1, and suggest that a principal role of RB1 in development is coordinating the events required for terminal differentiation.
Collapse
Affiliation(s)
- Ji Zhang
- a Department of Biochemistry ; St. Jude Children's Research Hospital ; Memphis , TN USA.,b Current address: Cancer Biology & Genetics; Memorial Sloan-Kettering Cancer Center ; New York , NY USA
| | - Melanie R Loyd
- a Department of Biochemistry ; St. Jude Children's Research Hospital ; Memphis , TN USA.,c Hartwell Center for Bioinformatics and Biotechnology; St. Jude Children's Research Hospital ; Memphis , TN USA
| | - Mindy S Randall
- a Department of Biochemistry ; St. Jude Children's Research Hospital ; Memphis , TN USA
| | - John J Morris
- c Hartwell Center for Bioinformatics and Biotechnology; St. Jude Children's Research Hospital ; Memphis , TN USA
| | - Jayesh G Shah
- d Cell & Molecular Biology; Lindsley F. Kimball Research Institute; New York Blood Center ; New York , NY USA
| | - Paul A Ney
- a Department of Biochemistry ; St. Jude Children's Research Hospital ; Memphis , TN USA.,d Cell & Molecular Biology; Lindsley F. Kimball Research Institute; New York Blood Center ; New York , NY USA.,e Current address: 1735 York Ave., New York , NY USA
| |
Collapse
|
31
|
Waugh KA, Leach SM, Moore BL, Bruno TC, Buhrman JD, Slansky JE. Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model. THE JOURNAL OF IMMUNOLOGY 2016; 197:1477-88. [PMID: 27371726 DOI: 10.4049/jimmunol.1600589] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/09/2016] [Indexed: 12/21/2022]
Abstract
Mechanisms of self-tolerance often result in CD8(+) tumor-infiltrating lymphocytes (TIL) with a hypofunctional phenotype incapable of tumor clearance. Using a transplantable colon carcinoma model, we found that CD8(+) T cells became tolerized in <24 h in an established tumor environment. To define the collective impact of pathways suppressing TIL function, we compared genome-wide mRNA expression of tumor-specific CD8(+) T cells from the tumor and periphery. Notably, gene expression induced during TIL hypofunction more closely resembled self-tolerance than viral exhaustion. Differential gene expression was refined to identify a core set of genes that defined hypofunctional TIL; these data comprise the first molecular profile of tumor-specific TIL that are naturally responding and represent a polyclonal repertoire. The molecular profile of TIL was further dissected to determine the extent of overlap and distinction between pathways that collectively restrict T cell functions. As suggested by the molecular profile of TIL, protein expression of inhibitory receptor LAG-3 was differentially regulated throughout prolonged late-G1/early-S phase of the cell cycle. Our data may accelerate efficient identification of combination therapies to boost anti-tumor function of TIL specifically against tumor cells.
Collapse
Affiliation(s)
| | - Sonia M Leach
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206
| | - Brandon L Moore
- University of Colorado School of Medicine, Aurora, CO 80045; and
| | - Tullia C Bruno
- University of Colorado School of Medicine, Aurora, CO 80045; and
| | | | - Jill E Slansky
- University of Colorado School of Medicine, Aurora, CO 80045; and
| |
Collapse
|
32
|
Abstract
The preimplantation development stage of mammalian embryogenesis consists of a series of highly conserved, regulated, and predictable cell divisions. This process is essential to allow the rapid expansion and differentiation of a single-cell zygote into a multicellular blastocyst containing cells of multiple developmental lineages. This period of development, also known as the germinal stage, encompasses several important developmental transitions, which are accompanied by dramatic changes in cell cycle profiles and dynamics. These changes are driven primarily by differences in the establishment and enforcement of cell cycle checkpoints, which must be bypassed to facilitate the completion of essential cell cycle events. Much of the current knowledge in this area has been amassed through the study of knockout models in mice. These mouse models are powerful experimental tools, which have allowed us to dissect the relative dependence of the early embryonic cell cycles on various aspects of the cell cycle machinery and highlight the extent of functional redundancy between members of the same gene family. This chapter will explore the ways in which the cell cycle machinery, their accessory proteins, and their stimuli operate during mammalian preimplantation using mouse models as a reference and how this allows for the usually well-defined stages of the cell cycle to be shaped and transformed during this unique and critical stage of development.
Collapse
|
33
|
Waugh KA, Leach SM, Slansky JE. Targeting Transcriptional Regulators of CD8+ T Cell Dysfunction to Boost Anti-Tumor Immunity. Vaccines (Basel) 2015; 3:771-802. [PMID: 26393659 PMCID: PMC4586477 DOI: 10.3390/vaccines3030771] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 02/07/2023] Open
Abstract
Transcription is a dynamic process influenced by the cellular environment: healthy, transformed, and otherwise. Genome-wide mRNA expression profiles reflect the collective impact of pathways modulating cell function under different conditions. In this review we focus on the transcriptional pathways that control tumor infiltrating CD8+ T cell (TIL) function. Simultaneous restraint of overlapping inhibitory pathways may confer TIL resistance to multiple mechanisms of suppression traditionally referred to as exhaustion, tolerance, or anergy. Although decades of work have laid a solid foundation of altered transcriptional networks underlying various subsets of hypofunctional or “dysfunctional” CD8+ T cells, an understanding of the relevance in TIL has just begun. With recent technological advances, it is now feasible to further elucidate and utilize these pathways in immunotherapy platforms that seek to increase TIL function.
Collapse
Affiliation(s)
- Katherine A Waugh
- University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA.
| | - Sonia M Leach
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA.
| | - Jill E Slansky
- University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA.
| |
Collapse
|
34
|
Iglesias-Ara A, Zenarruzabeitia O, Buelta L, Merino J, Zubiaga AM. E2F1 and E2F2 prevent replicative stress and subsequent p53-dependent organ involution. Cell Death Differ 2015; 22:1577-89. [PMID: 25656653 DOI: 10.1038/cdd.2015.4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 12/19/2014] [Accepted: 01/06/2015] [Indexed: 11/09/2022] Open
Abstract
Tissue homeostasis requires tight regulation of cellular proliferation, differentiation and apoptosis. E2F1 and E2F2 transcription factors share a critical role in tissue homeostasis, since their combined inactivation results in overall organ involution, specially affecting the pancreatic gland, which subsequently triggers diabetes. We have examined the mechanism by which these E2Fs regulate tissue homeostasis. We show that pancreas atrophy in E2F1/E2F2 double-knockout (DKO) mice is associated with mitochondrial apoptosis and activation of the p53 pathway in young animals, before the development of diabetes. A deregulated expression of E2F target genes was detected in pancreatic cells of young DKO animals, along with unscheduled DNA replication and activation of a DNA damage response. Importantly, suppression of DNA replication in vivo with aphidicolin led to a significant inhibition of the p53 pathway in DKO pancreas, implying a causal link between DNA replication stress and p53 activation in this model. We further show that activation of the p53 pathway has a key role in the aberrant phenotype of DKO mice, since targeted inactivation of p53 gene abrogated cellular apoptosis and prevented organ involution and insulin-dependent diabetes in mice lacking E2F1/E2F2. Unexpectedly, p53 inactivation unmasked oncogenic features of E2F1/E2F2-depleted cells, as evidenced by an accelerated tumor development in triple-knockout mice compared with p53(-/-) mice. Collectively, our data reveal a role for E2F1 and E2F2 as suppressors of replicative stress in differentiating cells, and uncover the existence of a robust E2F-p53 regulatory axis to enable tissue homeostasis and prevent tumorigenesis. These findings have implications in the design of approaches targeting E2F for cancer therapy.
Collapse
Affiliation(s)
- A Iglesias-Ara
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, Bilbao, Spain
| | - O Zenarruzabeitia
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, Bilbao, Spain
| | - L Buelta
- University of Cantabria-IDIVAL, Cantabria, Spain
| | - J Merino
- University of Cantabria-IDIVAL, Cantabria, Spain
| | - A M Zubiaga
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, Bilbao, Spain
| |
Collapse
|
35
|
Maldonado EN, Delgado I, Furland NE, Buqué X, Iglesias A, Aveldaño MI, Zubiaga A, Fresnedo O, Ochoa B. The E2F2 transcription factor sustains hepatic glycerophospholipid homeostasis in mice. PLoS One 2014; 9:e112620. [PMID: 25396754 PMCID: PMC4232400 DOI: 10.1371/journal.pone.0112620] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 10/09/2014] [Indexed: 12/29/2022] Open
Abstract
Increasing evidence links metabolic signals to cell proliferation, but the molecular wiring that connects the two core machineries remains largely unknown. E2Fs are master regulators of cellular proliferation. We have recently shown that E2F2 activity facilitates the completion of liver regeneration after partial hepatectomy (PH) by regulating the expression of genes required for S-phase entry. Our study also revealed that E2F2 determines the duration of hepatectomy-induced hepatic steatosis. A transcriptomic analysis of normal adult liver identified “lipid metabolism regulation” as a major E2F2 functional target, suggesting that E2F2 has a role in lipid homeostasis. Here we use wild-type (E2F2+/+) and E2F2 deficient (E2F2−/−) mice to investigate the in vivo role of E2F2 in the composition of liver lipids and fatty acids in two metabolically different contexts: quiescence and 48-h post-PH, when cellular proliferation and anabolic demands are maximal. We show that liver regeneration is accompanied by large triglyceride and protein increases without changes in total phospholipids both in E2F2+/+ and E2F2−/− mice. Remarkably, we found that the phenotype of quiescent liver tissue from E2F2−/− mice resembles the phenotype of proliferating E2F2+/+ liver tissue, characterized by a decreased phosphatidylcholine to phosphatidylethanolamine ratio and a reprogramming of genes involved in generation of choline and ethanolamine derivatives. The diversity of fatty acids in total lipid, triglycerides and phospholipids was essentially preserved on E2F2 loss both in proliferating and non-proliferating liver tissue, although notable exceptions in inflammation-related fatty acids of defined phospholipid classes were detected. Overall, our results indicate that E2F2 activity sustains the hepatic homeostasis of major membrane glycerolipid components while it is dispensable for storage glycerolipid balance.
Collapse
Affiliation(s)
- Eduardo N. Maldonado
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country, Leioa, Spain
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas y Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Igotz Delgado
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country, Leioa, Spain
| | - Natalia E. Furland
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas y Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Xabier Buqué
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country, Leioa, Spain
| | - Ainhoa Iglesias
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country, Leioa, Spain
| | - Marta I. Aveldaño
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Consejo Nacional de Investigaciones Científicas y Técnicas y Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Ana Zubiaga
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country, Leioa, Spain
| | - Olatz Fresnedo
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country, Leioa, Spain
| | - Begoña Ochoa
- Department of Physiology, Faculty of Medicine and Dentistry, University of the Basque Country, Leioa, Spain
- * E-mail:
| |
Collapse
|
36
|
Nakahata AM, Suzuki DE, Rodini CO, Fiuza ML, Okamoto OK. RNAi-mediated knockdown of E2F2 inhibits tumorigenicity of human glioblastoma cells. Oncol Lett 2014; 8:1487-1491. [PMID: 25202354 PMCID: PMC4156183 DOI: 10.3892/ol.2014.2369] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 06/19/2014] [Indexed: 02/06/2023] Open
Abstract
In a previous genome-wide expression profiling study, we identified E2F2 as a hyperexpressed gene in stem-like cells of distinct glioblastoma multiforme (GBM) specimens. Since the encoded E2F2 transcription factor has been implicated in both tumor suppression and tumor development, we conducted a functional study to investigate the pertinence of E2F2 to human gliomagenesis. E2F2 expression was knocked down by transfecting U87MG cells with plasmids carrying a specific silencing shRNA. Upon E2F2 silencing, in vitro cell proliferation was significantly reduced, as indicated by a time-course analysis of viable tumor cells. Anchorage-independent cell growth was also significantly inhibited after E2F2 silencing, based on cell colony formation in soft agar. Subcutaneous and orthotopic xenograft models of GBM in nude mice also indicated inhibition of tumor development in vivo, following E2F2 silencing. As expression of the E2F2 gene is associated with glioblastoma stem cells and is involved in the transformation of human astrocytes, the present findings suggest that E2F2 is involved in gliomagenesis and could be explored as a potential therapeutic target in malignant gliomas.
Collapse
Affiliation(s)
- Adriana M Nakahata
- Department of Neurology and Neurosurgery, Experimental Neurology Unit, Federal University of São Paulo, São Paulo 04023-900, Brazil ; Department of Genetics and Evolutionary Biology, Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| | - Daniela E Suzuki
- Department of Neurology and Neurosurgery, Experimental Neurology Unit, Federal University of São Paulo, São Paulo 04023-900, Brazil ; Department of Genetics and Evolutionary Biology, Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| | - Carolina O Rodini
- Department of Genetics and Evolutionary Biology, Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| | - Mayara L Fiuza
- Department of Genetics and Evolutionary Biology, Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| | - Oswaldo K Okamoto
- Department of Genetics and Evolutionary Biology, Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| |
Collapse
|
37
|
The E2F transcription factors regulate tumor development and metastasis in a mouse model of metastatic breast cancer. Mol Cell Biol 2014; 34:3229-43. [PMID: 24934442 DOI: 10.1128/mcb.00737-14] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
While the E2F transcription factors (E2Fs) have a clearly defined role in cell cycle control, recent work has uncovered new functions. Using genomic signature methods, we predicted a role for the activator E2F transcription factors in the mouse mammary tumor virus (MMTV)-polyomavirus middle T oncoprotein (PyMT) mouse model of metastatic breast cancer. To genetically test the hypothesis that the E2Fs function to regulate tumor development and metastasis, we interbred MMTV-PyMT mice with E2F1, E2F2, or E2F3 knockout mice. With the ablation of individual E2Fs, we noted alterations of tumor latency, histology, and vasculature. Interestingly, we noted striking reductions in metastatic capacity and in the number of circulating tumor cells in both the E2F1 and E2F2 knockout backgrounds. Investigating E2F target genes that mediate metastasis, we found that E2F loss led to decreased levels of vascular endothelial growth factor (Vegfa), Bmp4, Cyr61, Nupr1, Plod 2, P4ha1, Adamts1, Lgals3, and Angpt2. These gene expression changes indicate that the E2Fs control the expression of genes critical to angiogenesis, the remodeling of the extracellular matrix, tumor cell survival, and tumor cell interactions with vascular endothelial cells that facilitate metastasis to the lungs. Taken together, these results reveal that the E2F transcription factors play key roles in mediating tumor development and metastasis in addition to their well-characterized roles in cell cycle control.
Collapse
|
38
|
Cleveland SM, Smith S, Tripathi R, Mathias EM, Goodings C, Elliott N, Peng D, El-Rifai W, Yi D, Chen X, Li L, Mullighan C, Downing JR, Love P, Davé UP. Lmo2 induces hematopoietic stem cell-like features in T-cell progenitor cells prior to leukemia. Stem Cells 2014; 31:882-94. [PMID: 23378057 DOI: 10.1002/stem.1345] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 01/03/2013] [Indexed: 12/14/2022]
Abstract
LIM domain only 2 (Lmo2) is frequently deregulated in sporadic and gene therapy-induced acute T-cell lymphoblastic leukemia (T-ALL) where its overexpression is an important initiating mutational event. In transgenic and retroviral mouse models, Lmo2 expression can be enforced in multiple hematopoietic lineages but leukemia only arises from T cells. These data suggest that Lmo2 confers clonal growth advantage in T-cell progenitors. We analyzed proliferation, differentiation, and cell death in CD2-Lmo2 transgenic thymic progenitor cells to understand the cellular effects of enforced Lmo2 expression. Most impressively, Lmo2 transgenic T-cell progenitor cells were blocked in differentiation, quiescent, and immortalized in vitro on OP9-DL1 stromal cells. These cellular effects were concordant with a transcriptional signature in Lmo2 transgenic T-cell progenitor cells that is also present in hematopoietic stem cells (HSCs) and early T-cell precursor ALL. These results are significant in light of the crucial role of Lmo2 in the maintenance of the HSC. The cellular effects and transcriptional effects have implications for LMO2-dependent leukemogenesis and the treatment of LMO2-induced T-ALL.
Collapse
Affiliation(s)
- Susan M Cleveland
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee 37232-6307, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Andrechek ER. HER2/Neu tumorigenesis and metastasis is regulated by E2F activator transcription factors. Oncogene 2013; 34:217-25. [PMID: 24362522 PMCID: PMC4067469 DOI: 10.1038/onc.2013.540] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 10/30/2013] [Accepted: 11/05/2013] [Indexed: 02/06/2023]
Abstract
HER2 / Neu is amplified and overexpressed in a large proportion of human breast cancers, but the signaling pathways that contribute to tumor development and metastatic progression are not completely understood. Using gene expression data and pathway signatures we predicted a role for activator E2F transcription factors in Neu induced tumors. This was genetically tested by interbreeding Neu transgenics with knockouts of the three activator E2Fs. Loss of any E2F delayed Neu induced tumor onset. E2F1 loss accelerated tumor growth while E2F2 and E2F3 loss did not. Strikingly, it was observed that loss of E2F1 or E2F2 significantly reduced the metastatic capacity of the tumor and this was associated with a reduction in circulating tumor cells in the E2F2 knockout. Gene expression analysis between the tumors in the various E2F mutant backgrounds revealed that there was extensive compensation by other E2F family members in the individual knockouts, underscoring the importance of the E2Fs in HER2 / Neu induced tumors. Extension to HER2 positive human breast cancer revealed a number of HER2+ subtypes based on E2F activity with differences in relapse free survival times. Taken together these data demonstrate that the E2F transcription factors are integral to HER2+ tumor development and progression.
Collapse
Affiliation(s)
- E R Andrechek
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
40
|
Costa C, Paramio JM, Santos M. Skin Tumors Rb(eing) Uncovered. Front Oncol 2013; 3:307. [PMID: 24381932 PMCID: PMC3865458 DOI: 10.3389/fonc.2013.00307] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/04/2013] [Indexed: 11/23/2022] Open
Abstract
The Rb1 gene was the first bona fide tumor suppressor identified and cloned more than 25 years ago. Since then, a plethora of studies have revealed the functions of pRb and the existence of a sophisticated and strictly regulated pathway that modulates such functional roles. An emerging paradox affecting Rb1 in cancer connects the relatively low number of mutations affecting Rb1 gene in specific human tumors, compared with the widely functional inactivation of pRb in most, if not in all, human cancers. The existence of a retinoblastoma family of proteins pRb, p107, and p130 and their potential unique and overlapping functions as master regulators of cell cycle progression and transcriptional modulation by similar processes, may provide potential clues to explain such conundrum. Here, we will review the development of different genetically engineered mouse models, in particular those affecting stratified epithelia, and how they have offered new avenues to understand the roles of the Rb family members and their targets in the context of tumor development and progression.
Collapse
Affiliation(s)
- Clotilde Costa
- Molecular Oncology Unit, Department of Basic Research, Centro de Investigaciones Energéticas Medioambientales y Teconológicas (ed70A) , Madrid , Spain
| | - Jesús M Paramio
- Molecular Oncology Unit, Department of Basic Research, Centro de Investigaciones Energéticas Medioambientales y Teconológicas (ed70A) , Madrid , Spain
| | - Mirentxu Santos
- Molecular Oncology Unit, Department of Basic Research, Centro de Investigaciones Energéticas Medioambientales y Teconológicas (ed70A) , Madrid , Spain
| |
Collapse
|
41
|
Laresgoiti U, Apraiz A, Olea M, Mitxelena J, Osinalde N, Rodriguez JA, Fullaondo A, Zubiaga AM. E2F2 and CREB cooperatively regulate transcriptional activity of cell cycle genes. Nucleic Acids Res 2013; 41:10185-98. [PMID: 24038359 PMCID: PMC3905855 DOI: 10.1093/nar/gkt821] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
E2F2 is essential for the maintenance of T lymphocyte quiescence. To identify the full set of E2F2 target genes, and to gain further understanding of the role of E2F2 in transcriptional regulation, we have performed ChIP-chip analyses across the genome of lymph node–derived T lymphocytes. Here we show that during quiescence, E2F2 binds the promoters of a large number of genes involved in DNA metabolism and cell cycle regulation, concomitant with their transcriptional silencing. A comparison of ChIP-chip data with expression profiling data on resting E2f2−/− T lymphocytes identified a subset of 51 E2F2-specific target genes, most of which are upregulated on E2F2 loss. Luciferase reporter assays showed a retinoblastoma-independent role for E2F2 in the negative regulation of these target genes. Importantly, we show that the DNA binding activity of the transcription factor CREB contributes to E2F2-mediated repression of Mcm5 and Chk1 promoters. siRNA-mediated CREB knockdown, expression of a dominant negative KCREB mutant or disruption of CREB binding by mutating a CRE motif on Mcm5 promoter, relieved E2F2-mediated transcriptional repression. Taken together, our data uncover a new regulatory mechanism for E2F-mediated transcriptional control, whereby E2F2 and CREB cooperate in the transcriptional repression of a subset of E2F2 target genes.
Collapse
Affiliation(s)
- Usua Laresgoiti
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, UPV/EHU, Bilbao 48940, Spain and Department of Biochemistry and Molecular Biology, University of the Basque Country, UPV/EHU, Bilbao 48940, Spain
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Contrasting roles of E2F2 and E2F3 in cardiac neovascularization. PLoS One 2013; 8:e65755. [PMID: 23799044 PMCID: PMC3683051 DOI: 10.1371/journal.pone.0065755] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 04/29/2013] [Indexed: 01/23/2023] Open
Abstract
Insufficient neovascularization, characterized by poor endothelial cell (EC) growth, contributes to the pathogenesis of ischemic heart disease and limits cardiac tissue preservation and regeneration. The E2F family of transcription factors are critical regulators of the genes responsible for cell-cycle progression and growth; however, the specific roles of individual E2Fs in ECs are not well understood. Here we investigated the roles of E2F2 and E2F3 in EC growth, angiogenesis, and their functional impact on myocardial infarction (MI). An endothelial-specific E2F3-deficient mouse strain VE-Cre; E2F3(fl/fl) was generated, and MI was surgically induced in VE-Cre; E2F3(fl/fl) and E2F2-null (E2F2 KO) mice and their wild-type (WT) littermates, VE-Cre; E2F3(+/+) and E2F2 WT, respectively. The cardiac function, infarct size, and vascular density were significantly better in E2F2 KO mice and significantly worse in VE-Cre; E2F3(fl/fl) mice than in their WT littermates. The loss of E2F2 expression was associated with an increase in the proliferation of ECs both in vivo and in vitro, while the loss of E2F3 expression led to declines in EC proliferation. Thus, E2F3 promotes while E2F2 suppresses ischemic cardiac repair through corresponding changes in EC proliferation; and differential targeting of specific E2F members may provide a novel strategy for therapeutic angiogenesis of ischemic heart disease.
Collapse
|
43
|
Osinalde N, Olea M, Mitxelena J, Aloria K, Rodriguez JA, Fullaondo A, Arizmendi JM, Zubiaga AM. The nuclear protein ALY binds to and modulates the activity of transcription factor E2F2. Mol Cell Proteomics 2013; 12:1087-98. [PMID: 23297349 DOI: 10.1074/mcp.m112.024158] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
E2F transcription factors control the expression of genes involved in a variety of essential cellular processes and consequently their activity needs to be tightly regulated. Protein-protein interactions are thought to be key modulators of E2F activity. To gain insight into the mechanisms that regulate the activity of E2F2, we searched for novel proteins that associate with this transcription factor. We show that the nuclear protein ALY (THO complex 4), originally described as a transcriptional co-activator, associates with DNA-bound E2F2 and represses its transcriptional activity. The capacity of ALY to modulate gene expression was analyzed with expression microarrays by characterizing the transcriptome of E2F2 expressing HEK293T cells in which ALY was either overexpressed or silenced. We show that ALY influences the expression of more than 400 genes, including 98 genes bearing consensus E2F motifs. Thus, ALY emerges as a novel E2F2-interacting protein and a relevant modulator of E2F-responsive gene expression.
Collapse
Affiliation(s)
- Nerea Osinalde
- Department of Biochemistry and Molecular Biology, University of the Basque Country, UPV/EHU, 48940 Leioa, Spain
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Chen HZ, Ouseph MM, Li J, Pécot T, Chokshi V, Kent L, Bae S, Byrne M, Duran C, Comstock G, Trikha P, Mair M, Senapati S, Martin CK, Gandhi S, Wilson N, Liu B, Huang YW, Thompson JC, Raman S, Singh S, Leone M, Machiraju R, Huang K, Mo X, Fernandez S, Kalaszczynska I, Wolgemuth DJ, Sicinski P, Huang T, Jin V, Leone G. Canonical and atypical E2Fs regulate the mammalian endocycle. Nat Cell Biol 2012; 14:1192-202. [PMID: 23064266 PMCID: PMC3616487 DOI: 10.1038/ncb2595] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 09/03/2012] [Indexed: 02/06/2023]
Abstract
The endocycle is a variant cell cycle consisting of successive DNA synthesis and Gap phases that yield highly polyploid cells. Although essential for metazoan development, relatively little is known about its control or physiologic role in mammals. Using novel lineage-specific cre mice we identified two opposing arms of the E2F program, one driven by canonical transcription activation (E2F1, E2F2 and E2F3) and the other by atypical repression (E2F7 and E2F8), that converge on the regulation of endocycles in vivo. Ablation of canonical activators in the two endocycling tissues of mammals, trophoblast giant cells in the placenta and hepatocytes in the liver, augmented genome ploidy, whereas ablation of atypical repressors diminished ploidy. These two antagonistic arms coordinate the expression of a unique G2/M transcriptional program that is critical for mitosis, karyokinesis and cytokinesis. These results provide in vivo evidence for a direct role of E2F family members in regulating non-traditional cell cycles in mammals.
Collapse
Affiliation(s)
- Hui-Zi Chen
- Solid Tumor Biology Program, Department of Molecular Virology, Immunology and Medical Genetics, Department of Molecular Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Matsumoto A, Nakayama KI. Role of key regulators of the cell cycle in maintenance of hematopoietic stem cells. Biochim Biophys Acta Gen Subj 2012; 1830:2335-44. [PMID: 22820018 DOI: 10.1016/j.bbagen.2012.07.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 06/26/2012] [Accepted: 07/10/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND Hematopoietic stem cells (HSCs) are characterized by pluripotentiality and self-renewal ability. To maintain a supply of mature blood cells and to avoid HSC exhaustion during the life span of an organism, most HSCs remain quiescent, with only a limited number entering the cell cycle. SCOPE OF REVIEW The molecular mechanisms by which quiescence is maintained in HSCs are addressed, with recent genetic studies having provided important insight into the relation between the cell cycle activity and stemness of HSCs. MAJOR CONCLUSIONS The cell cycle is tightly regulated in HSCs by complex factors. Key regulators of the cell cycle in other cell types-including cyclins, cyclin-dependent kinases (CDKs), the retinoblastoma protein family, the transcription factor E2F, and CDK inhibitors-also contribute to such regulation in HSCs. Most, but not all, of these regulators are necessary for maintenance of HSCs, with abnormal activation or suppression of the cell cycle resulting in HSC exhaustion. The cell cycle in HSCs is also regulated by external factors such as cytokines produced by niche cells as well as by the ubiquitin-proteasome pathway. GENERAL SIGNIFICANCE Studies of the cell cycle in HSCs may shed light on the pathogenesis of hematopoietic disorders, serve as a basis for the development of new therapeutic strategies for such disorders, prove useful for the expansion of HSCs in vitro as a possible replacement for blood transfusion, and provide insight into stem cell biology in general. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- Akinobu Matsumoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | | |
Collapse
|
46
|
Ouseph MM, Li J, Chen HZ, Pécot T, Wenzel P, Thompson JC, Comstock G, Chokshi V, Byrne M, Forde B, Chong JL, Huang K, Machiraju R, de Bruin A, Leone G. Atypical E2F repressors and activators coordinate placental development. Dev Cell 2012; 22:849-62. [PMID: 22516201 DOI: 10.1016/j.devcel.2012.01.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2011] [Revised: 11/23/2011] [Accepted: 01/18/2012] [Indexed: 02/06/2023]
Abstract
The evolutionarily ancient arm of the E2f family of transcription factors consisting of the two atypical members E2f7 and E2f8 is essential for murine embryonic development. However, the critical tissues, cellular processes, and molecular pathways regulated by these two factors remain unknown. Using a series of fetal and placental lineage-specific cre mice, we show that E2F7/E2F8 functions in extraembryonic trophoblast lineages are both necessary and sufficient to carry fetuses to term. Expression profiling and biochemical approaches exposed the canonical E2F3a activator as a key family member that antagonizes E2F7/E2F8 functions. Remarkably, the concomitant loss of E2f3a normalized placental gene expression programs, corrected placental defects, and fostered the survival of E2f7/E2f8-deficient embryos to birth. In summary, we identified a placental transcriptional network tightly coordinated by activation and repression through two distinct arms of the E2F family that is essential for extraembryonic cell proliferation, placental development, and fetal viability.
Collapse
Affiliation(s)
- Madhu M Ouseph
- Solid Tumor Biology Program, Department of Molecular Virology, Immunology and Medical Genetics, Human Cancer Genetics Program, Comprehensive Cancer Center, College of Medicine and Public Health, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Ito T, Maeda T, Senpuku H. Roles of salivary components in Streptococcus mutans colonization in a new animal model using NOD/SCID.e2f1-/- mice. PLoS One 2012; 7:e32063. [PMID: 22363797 PMCID: PMC3283720 DOI: 10.1371/journal.pone.0032063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 01/20/2012] [Indexed: 01/28/2023] Open
Abstract
Streptococcus mutans plays an important role in biofilm formation on the tooth surface and is the primary causative agent of dental caries. The binding of S. mutans to the salivary pellicle is of considerable etiologic significance and is important in biofilm development. Recently, we produced NOD/SCID.e2f1(-/-) mice that show hyposalivation, lower salivary antibody, and an extended life span compared to the parent strain: NOD.e2f1(-/-). In this study we used NOD/SCID.e2f1(-/-) 4 or 6 mice to determine the roles of several salivary components in S. mutans colonization in vivo. S. mutans colonization in NOD/SCID.e2f1(-/-) mice was significantly increased when mice were pre-treated with human saliva or commercial salivary components. Interestingly, pre-treatment with secretory IgA (sIgA) at physiological concentrations promoted significant colonization of S. mutans compared with sIgA at higher concentrations, or with human saliva or other components. Our data suggest the principal effects of specific sIgA on S. mutans occur during S. mutans colonization, where the appropriate concentration of specific sIgA may serve as an anti-microbial agent, agglutinin, or an adherence receptor to surface antigens. Further, specific sIgA supported biofilm formation when the mice were supplied 1% sucrose water and a non-sucrose diet. The data suggests that there are multiple effects exerted by sIgA in S. mutans colonization, with synergistic effects evident under the condition of sIgA and limited nutrients on colonization in NOD/SCID.e2f1(-/-) mice. This is a new animal model that can be used to assess prevention methods for dental biofilm-dependent diseases such as dental caries.
Collapse
Affiliation(s)
- Tatsuro Ito
- Department of Pediatric Dentistry, Nihon University Graduate School of Dentistry at Matsudo, Chiba, Japan
- Department of Bacteriology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takahide Maeda
- Department of Pediatric Dentistry, Nihon University Graduate School of Dentistry at Matsudo, Chiba, Japan
| | - Hidenobu Senpuku
- Department of Bacteriology, National Institute of Infectious Diseases, Tokyo, Japan
- * E-mail:
| |
Collapse
|
48
|
Schietinger A, Delrow JJ, Basom RS, Blattman JN, Greenberg PD. Rescued tolerant CD8 T cells are preprogrammed to reestablish the tolerant state. Science 2012; 335:723-7. [PMID: 22267581 DOI: 10.1126/science.1214277] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tolerant self-antigen-specific CD8 T cells fail to proliferate in response to antigen, thereby preventing autoimmune disease. By using an in vivo mouse model, we show that tolerant T cells proliferate and become functional under lymphopenic conditions, even in a tolerogenic environment. However, T cell rescue is only transient, with tolerance reimposed upon lymphorepletion even in the absence of tolerogen (self-antigen), challenging the prevailing paradigm that continuous antigen exposure is critical to maintain tolerance. Genome-wide messenger RNA and microRNA profiling revealed that tolerant T cells have a tolerance-specific gene profile that can be temporarily overridden under lymphopenic conditions but is inevitably reimposed, which suggests epigenetic regulation. These insights into the regulatory mechanisms that maintain or break self-tolerance may lead to new strategies for the treatment of cancer and autoimmunity.
Collapse
Affiliation(s)
- Andrea Schietinger
- Department of Immunology, University of Washington (UW), Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
49
|
Abstract
Mouse models of lupus have for many years provided accessible and reliable research systems for the pathogenesis and therapy of systemic autoimmune disease, spanning a spectrum of inbred strains that develop spontaneous disease to experimentally induced, sometimes genetically manipulated animals. Nearly all the models share in common the development of glomerulonephritis and autoantibodies, including antinuclear and DNA specificities, the most common endpoints examined in experimental studies, but exhibit specific differences in the incidence of other end-organ manifestations such as hemolytic anemia, arthritis, dermatitis, and vasculitis. This chapter contrasts the clinical characteristics of these various models, providing an outline for their use and analysis.
Collapse
Affiliation(s)
- Stanford L Peng
- Rheumatology Clinical Research Unit, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| |
Collapse
|
50
|
Tajouri L, Fernandez F, Griffiths LR. Gene expression studies in multiple sclerosis. Curr Genomics 2011; 8:181-9. [PMID: 18645602 DOI: 10.2174/138920207780833829] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Revised: 02/14/2007] [Accepted: 03/14/2007] [Indexed: 11/22/2022] Open
Abstract
Multiple sclerosis (MS) is a serious neurological disorder affecting young Caucasian individuals, usually with an age of onset at 18 to 40 years old. Females account for approximately 60x of MS cases and the manifestation and course of the disease is highly variable from patient to patient. The disorder is characterised by the development of plaques within the central nervous system (CNS). Many gene expression studies have been undertaken to look at the specific patterns of gene transcript levels in MS. Human tissues and experimental mice were used in these gene-profiling studies and a very valuable and interesting set of data has resulted from these various expression studies. In general, genes showing variable expression include mainly immunological and inflammatory genes, stress and antioxidant genes, as well as metabolic and central nervous system markers. Of particular interest are a number of genes localised to susceptible loci previously shown to be in linkage with MS. However due to the clinical complexity of the disease, the heterogeneity of the tissues used in expression studies, as well as the variable DNA chips/membranes used for the gene profiling, it is difficult to interpret the available information. Although this information is essential for the understanding of the pathogenesis of MS, it is difficult to decipher and define the gene pathways involved in the disorder. Experiments in gene expression profiling in MS have been numerous and lists of candidates are now available for analysis. Researchers have investigated gene expression in peripheral mononuclear white blood cells (PBMCs), in MS animal models Experimental Allergic Encephalomyelitis (EAE) and post mortem MS brain tissues. This review will focus on the results of these studies.
Collapse
Affiliation(s)
- Lotti Tajouri
- Genomics Research Centre, School of Medical Science, Griffith University Gold Coast, Southport, Queensland, 4215 Australia
| | | | | |
Collapse
|