1
|
Huang A, Rao J, Feng X, Li X, Xu T, Yao L. Breaking New Ground: Unraveling the USP1/ID3/E12/P21 Axis in Vascular Calcification. Transl Res 2024:S1931-5244(24)00170-1. [PMID: 39326697 DOI: 10.1016/j.trsl.2024.09.002] [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: 05/05/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
Vascular calcification (VC) poses significant challenges in cardiovascular health. This study employs single-cell transcriptome sequencing to dissect cellular dynamics in this process. We identify distinct cell subgroups, notably in vascular smooth muscle cells (VSMCs), and observe differences between calcified atherosclerotic cores and adjacent regions. Further exploration reveals ID3 as a key gene regulating VSMC function. In vitro experiments demonstrate ID3's interaction with USP1 and E12, modulating cell proliferation and osteogenic differentiation. Animal models confirm the critical role of the USP1/ID3/E12/P21 axis in VC. This study sheds light on a novel regulatory mechanism, offering potential therapeutic targets.
Collapse
Affiliation(s)
- Aoran Huang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China
| | - Jianyun Rao
- Outpatient Management Office, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Xin Feng
- Department of Nephrology, Liaoning electric power central hospital, Shenyang 110000, China
| | - Xingru Li
- Department of Nephrology, Liaoning electric power central hospital, Shenyang 110000, China
| | - Tianhua Xu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China.
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, China.
| |
Collapse
|
2
|
Mihai A, Lee SY, Shinton S, Parker MI, Contreras AV, Zhang B, Rhodes M, Dunbrack RL, Zúñiga-Pflücker JC, Ciofani M, Zhuang Y, Wiest DL. E proteins control the development of NKγδT cells through their invariant T cell receptor. Nat Commun 2024; 15:5078. [PMID: 38871720 PMCID: PMC11176164 DOI: 10.1038/s41467-024-49496-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024] Open
Abstract
T cell receptor (TCR) signaling regulates important developmental transitions, partly through induction of the E protein antagonist, Id3. Although normal γδ T cell development depends on Id3, Id3 deficiency produces different phenotypes in distinct γδ T cell subsets. Here, we show that Id3 deficiency impairs development of the Vγ3+ subset, while markedly enhancing development of NKγδT cells expressing the invariant Vγ1Vδ6.3 TCR. These effects result from Id3 regulating both the generation of the Vγ1Vδ6.3 TCR and its capacity to support development. Indeed, the Trav15 segment, which encodes the Vδ6.3 TCR subunit, is directly bound by E proteins that control its expression. Once expressed, the Vγ1Vδ6.3 TCR specifies the innate-like NKγδT cell fate, even in progenitors beyond the normally permissive perinatal window, and this is enhanced by Id3-deficiency. These data indicate that the paradoxical behavior of NKγδT cells in Id3-deficient mice is determined by its stereotypic Vγ1Vδ6.3 TCR complex.
Collapse
Affiliation(s)
- Ariana Mihai
- Immunology Department, Duke University, Durham, NC, USA
| | - Sang-Yun Lee
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Susan Shinton
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Mitchell I Parker
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - Baojun Zhang
- Immunology Department, Duke University, Durham, NC, USA
| | - Michele Rhodes
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Roland L Dunbrack
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | - Maria Ciofani
- Immunology Department, Duke University, Durham, NC, USA
| | - Yuan Zhuang
- Immunology Department, Duke University, Durham, NC, USA
| | - David L Wiest
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Deng Z, Loyher PL, Lazarov T, Li L, Shen Z, Bhinder B, Yang H, Zhong Y, Alberdi A, Massague J, Sun JC, Benezra R, Glass CK, Elemento O, Iacobuzio-Donahue CA, Geissmann F. The nuclear factor ID3 endows macrophages with a potent anti-tumour activity. Nature 2024; 626:864-873. [PMID: 38326607 PMCID: PMC10881399 DOI: 10.1038/s41586-023-06950-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 12/07/2023] [Indexed: 02/09/2024]
Abstract
Macrophage activation is controlled by a balance between activating and inhibitory receptors1-7, which protect normal tissues from excessive damage during infection8,9 but promote tumour growth and metastasis in cancer7,10. Here we report that the Kupffer cell lineage-determining factor ID3 controls this balance and selectively endows Kupffer cells with the ability to phagocytose live tumour cells and orchestrate the recruitment, proliferation and activation of natural killer and CD8 T lymphoid effector cells in the liver to restrict the growth of a variety of tumours. ID3 shifts the macrophage inhibitory/activating receptor balance to promote the phagocytic and lymphoid response, at least in part by buffering the binding of the transcription factors ELK1 and E2A at the SIRPA locus. Furthermore, loss- and gain-of-function experiments demonstrate that ID3 is sufficient to confer this potent anti-tumour activity to mouse bone-marrow-derived macrophages and human induced pluripotent stem-cell-derived macrophages. Expression of ID3 is therefore necessary and sufficient to endow macrophages with the ability to form an efficient anti-tumour niche, which could be harnessed for cell therapy in cancer.
Collapse
Affiliation(s)
- Zihou Deng
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pierre-Louis Loyher
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tomi Lazarov
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Li Li
- Graduate Center, City University of New York, New York, NY, USA
| | - Zeyang Shen
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Bhavneet Bhinder
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell, New York, NY, USA
| | - Hairu Yang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yi Zhong
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Araitz Alberdi
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joan Massague
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joseph C Sun
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert Benezra
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher K Glass
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell, New York, NY, USA
| | | | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
4
|
Urata S, Yamaguchi S, Nambu A, Sudo K, Nakae S, Yasuda J. The roles of BST-2 in murine B cell development and on virus propagation. Microbiol Immunol 2023; 67:105-113. [PMID: 36604771 DOI: 10.1111/1348-0421.13049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/13/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
The bone marrow (BM) stromal cell antigen-2 (BST-2), also known as tetherin, CD317, PDCA-1, or HM1.24, is a membrane protein overexpressed in several types of tumors and may act as a promising target for cancer treatment via antibody-dependent cellular cytotoxicity. BST-2 is also expressed in human BM stromal cells (BMSC), which support B cell development. While the activity of BST-2 as an antiviral factor has been demonstrated, the expression patterns and the role of BST-2 on B-cell development and activation have not been investigated, especially in vivo. In this study, Bst2 knockout (Bst2-/- ) mice were generated to assess the role of BST-2 on B cell development and activation. It was observed that BST-2 was not expressed in BMSC or all B cell progenitors even in wild-type mice and does not play a significant role in B cell development. In addition, the loss of BST-2 had no effect on B cell activation. Furthermore and in contrast to the well-known antiviral role of BST-2, infection of vesicular stomatitis Indiana virus to the BM cells collected from the Bst2-/- mice produced less infectious virus compared with that from the WT mice. These results suggest that murine BST-2 is different from human BST-2 in the expression pattern, physiological function, in vivo, and might possess positive role on VSV replication.
Collapse
Affiliation(s)
- Shuzo Urata
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, Nagasaki, Japan
| | - Sachiko Yamaguchi
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Aya Nambu
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Katsuko Sudo
- Pre-clinical Research Center, Tokyo Medical University, Tokyo, Japan
| | - Susumu Nakae
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama, Japan
| | - Jiro Yasuda
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, Nagasaki, Japan.,Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
5
|
Singh S, Sarkar T, Jakubison B, Gadomski S, Spradlin A, Gudmundsson KO, Keller JR. Inhibitor of DNA binding proteins revealed as orchestrators of steady state, stress and malignant hematopoiesis. Front Immunol 2022; 13:934624. [PMID: 35990659 PMCID: PMC9389078 DOI: 10.3389/fimmu.2022.934624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/12/2022] [Indexed: 11/24/2022] Open
Abstract
Adult mammalian hematopoiesis is a dynamic cellular process that provides a continuous supply of myeloid, lymphoid, erythroid/megakaryocyte cells for host survival. This process is sustained by regulating hematopoietic stem cells (HSCs) quiescence, proliferation and activation under homeostasis and stress, and regulating the proliferation and differentiation of downstream multipotent progenitor (MPP) and more committed progenitor cells. Inhibitor of DNA binding (ID) proteins are small helix-loop-helix (HLH) proteins that lack a basic (b) DNA binding domain present in other family members, and function as dominant-negative regulators of other bHLH proteins (E proteins) by inhibiting their transcriptional activity. ID proteins are required for normal T cell, B cell, NK and innate lymphoid cells, dendritic cell, and myeloid cell differentiation and development. However, recent evidence suggests that ID proteins are important regulators of normal and leukemic hematopoietic stem and progenitor cells (HSPCs). This chapter will review our current understanding of the function of ID proteins in HSPC development and highlight future areas of scientific investigation.
Collapse
Affiliation(s)
- Shweta Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Tanmoy Sarkar
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Brad Jakubison
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Stephen Gadomski
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Andrew Spradlin
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Kristbjorn O. Gudmundsson
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Jonathan R. Keller
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
- *Correspondence: Jonathan R. Keller,
| |
Collapse
|
6
|
Ahmadi SE, Rahimi S, Zarandi B, Chegeni R, Safa M. MYC: a multipurpose oncogene with prognostic and therapeutic implications in blood malignancies. J Hematol Oncol 2021; 14:121. [PMID: 34372899 PMCID: PMC8351444 DOI: 10.1186/s13045-021-01111-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/12/2021] [Indexed: 12/17/2022] Open
Abstract
MYC oncogene is a transcription factor with a wide array of functions affecting cellular activities such as cell cycle, apoptosis, DNA damage response, and hematopoiesis. Due to the multi-functionality of MYC, its expression is regulated at multiple levels. Deregulation of this oncogene can give rise to a variety of cancers. In this review, MYC regulation and the mechanisms by which MYC adjusts cellular functions and its implication in hematologic malignancies are summarized. Further, we also discuss potential inhibitors of MYC that could be beneficial for treating hematologic malignancies.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Rahimi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahman Zarandi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rouzbeh Chegeni
- Medical Laboratory Sciences Program, College of Health and Human Sciences, Northern Illinois University, DeKalb, IL, USA.
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Zheng Q, Zheng T, Zhang A, Yan B, Li B, Zhang Z, Zhang Y. Hearing Loss in Id1 -/-; Id3 +/- and Id1 +/-; Id3 -/- Mice Is Associated With a High Incidence of Middle Ear Infection (Otitis Media). Front Genet 2021; 12:508750. [PMID: 34434211 PMCID: PMC8381378 DOI: 10.3389/fgene.2021.508750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 07/06/2021] [Indexed: 11/13/2022] Open
Abstract
Inhibitors of differentiation/DNA binding (Id) proteins are crucial for inner ear development, but whether Id mutations affect middle ear function remains unknown. In this study, we obtained Id1-/-; Id3+/- mice and Id1+/-; Id3-/- mice and carefully examined their middle ear morphology and auditory function. Our study revealed a high incidence (>50%) of middle ear infection in the compound mutant mice. These mutant mice demonstrated hearing impairment starting around 30 days of age, as the mutant mice presented elevated auditory brainstem response (ABR) thresholds compared to those of the littermate controls. The distortion product of otoacoustic emission (DPOAE) was also used to evaluate the conductive function of the middle ear, and we found much lower DPOAE amplitudes in the mutant mice, suggesting sound transduction in the mutant middle ear is compromised. This is the first study of the middle ears of Id compound mutant mice, and high incidence of middle ear infection determined by otoscopy and histological analysis of middle ear suggests that Id1/Id3 compound mutant mice are a novel model for human otitis media (OM).
Collapse
Affiliation(s)
- Qingyin Zheng
- Department of Otolaryngology – Head and Neck Surgery, Second Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, China
- School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Tihua Zheng
- Department of Otolaryngology – Head and Neck Surgery, Second Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, China
- College of Special Education, Hearing and Speech Rehabilitation Institute, Binzhou Medical University, Yantai, China
| | - Aizhen Zhang
- School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Bin Yan
- College of Special Education, Hearing and Speech Rehabilitation Institute, Binzhou Medical University, Yantai, China
| | - Bo Li
- College of Special Education, Hearing and Speech Rehabilitation Institute, Binzhou Medical University, Yantai, China
| | - Zhaoqiang Zhang
- School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Yan Zhang
- Department of Otolaryngology – Head and Neck Surgery, Second Affiliated Hospital of Xi’an Jiaotong University School of Medicine, Xi’an, China
| |
Collapse
|
8
|
He Y, Chen Q, Dai J, Cui Y, Zhang C, Wen X, Li J, Xiao Y, Peng X, Liu M, Shen B, Sha J, Hu Z, Li J, Shu W. Single-cell RNA-Seq reveals a highly coordinated transcriptional program in mouse germ cells during primordial follicle formation. Aging Cell 2021; 20:e13424. [PMID: 34174788 PMCID: PMC8282241 DOI: 10.1111/acel.13424] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 06/08/2021] [Indexed: 01/18/2023] Open
Abstract
The assembly of primordial follicles in mammals represents one of the most critical processes in ovarian biology. It directly affects the number of oocytes available to a female throughout her reproductive life. Premature depletion of primordial follicles contributes to the ovarian pathology primary ovarian insufficiency (POI). To delineate the developmental trajectory and regulatory mechanisms of oocytes during the process, we performed RNA‐seq on single germ cells from newborn (P0.5) ovaries. Three cell clusters were classified which corresponded to three cell states (germ cell cyst, cyst breakdown, and follicle) in the newborn ovary. By Monocle analysis, a uniform trajectory of oocyte development was built with a series of genes showed dynamic changes along the pseudo‐timeline. Gene Ontology term enrichment revealed a significant decrease in meiosis‐related genes and a dramatic increase in oocyte‐specific genes which marked the transition from a germ cell to a functional oocyte. We then established a network of regulons by using single‐cell regulatory network inference and clustering (SCENIC) algorithm and identified possible candidate transcription factors that may maintain transcription programs during follicle formation. Following functional studies further revealed the differential regulation of the identified regulon Id2 and its family member Id1, on the establishment of primordial follicle pool by using siRNA knockdown and genetic modified mouse models. In summary, our study systematically reconstructed molecular cascades in oocytes and identified a series of genes and molecular pathways in follicle formation and development.
Collapse
Affiliation(s)
- Yuanlin He
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
- Department of Epidemiology and Biostatistics International Joint Research Center on Environment and Human Health Center for Global Health School of Public Health Nanjing Medical University Nanjing China
| | - Qiuzhen Chen
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
- Department of Biotechnology Beijing Institute of Radiation Medicine Beijing China
- Computer School University of South China Hengyang China
| | - Juncheng Dai
- Department of Epidemiology and Biostatistics International Joint Research Center on Environment and Human Health Center for Global Health School of Public Health Nanjing Medical University Nanjing China
| | - Yiqiang Cui
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Chi Zhang
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Xidong Wen
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Jiazhao Li
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Yue Xiao
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Xiaoxu Peng
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Mingxi Liu
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Bin Shen
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
- Department of Epidemiology and Biostatistics International Joint Research Center on Environment and Human Health Center for Global Health School of Public Health Nanjing Medical University Nanjing China
| | - Jing Li
- State Key Laboratory of Reproductive Medicine Nanjing Medical University Nanjing China
| | - Wenjie Shu
- Department of Biotechnology Beijing Institute of Radiation Medicine Beijing China
| |
Collapse
|
9
|
Leong SP, Witz IP, Sagi-Assif O, Izraely S, Sleeman J, Piening B, Fox BA, Bifulco CB, Martini R, Newman L, Davis M, Sanders LM, Haussler D, Vaske OM, Witte M. Cancer microenvironment and genomics: evolution in process. Clin Exp Metastasis 2021; 39:85-99. [PMID: 33970362 DOI: 10.1007/s10585-021-10097-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023]
Abstract
Cancer heterogeneity is a result of genetic mutations within the cancer cells. Their proliferation is not only driven by autocrine functions but also under the influence of cancer microenvironment, which consists of normal stromal cells such as infiltrating immune cells, cancer-associated fibroblasts, endothelial cells, pericytes, vascular and lymphatic channels. The relationship between cancer cells and cancer microenvironment is a critical one and we are just on the verge to understand it on a molecular level. Cancer microenvironment may serve as a selective force to modulate cancer cells to allow them to evolve into more aggressive clones with ability to invade the lymphatic or vascular channels to spread to regional lymph nodes and distant sites. It is important to understand these steps of cancer evolution within the cancer microenvironment towards invasion so that therapeutic strategies can be developed to control or stop these processes.
Collapse
Affiliation(s)
- Stanley P Leong
- California Pacific Medical Center and Research Institute, San Francisco, USA
| | - Isaac P Witz
- The Shmunis School of Biomedicine and Cancer Research, School of Molecular Cell Biology & Biotechnology, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Orit Sagi-Assif
- The Shmunis School of Biomedicine and Cancer Research, School of Molecular Cell Biology & Biotechnology, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Sivan Izraely
- The Shmunis School of Biomedicine and Cancer Research, School of Molecular Cell Biology & Biotechnology, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Jonathan Sleeman
- European Center for Angioscience, Medizinische Fakultät Mannheim der Universität Heidelberg, Heidelberg, Germany
| | | | | | | | - Rachel Martini
- Department of Surgery, Weill Cornell Medical College, New York City, NY, USA.,Department of Genetics, University of Georgia, Athens, GA, USA
| | - Lisa Newman
- Department of Surgery, Weill Cornell Medical College, New York City, NY, USA
| | - Melissa Davis
- Department of Surgery, Weill Cornell Medical College, New York City, NY, USA.
| | - Lauren M Sanders
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz and UC Santa Cruz Genomics Institute, Santa Cruz, USA
| | - David Haussler
- UC Santa Cruz Genomics Institute and Howard Hughes Medical Institute, University of California Santa Cruz, Santa Cruz, USA.
| | - Olena M Vaske
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz and UC Santa Cruz Genomics Institute, Santa Cruz, USA
| | - Marlys Witte
- Department of Surgery, Neurosurgery and Pediatrics, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| |
Collapse
|
10
|
Point Mutation Specific Antibodies in B-Cell and T-Cell Lymphomas and Leukemias: Targeting IDH2, KRAS, BRAF and Other Biomarkers RHOA, IRF8, MYD88, ID3, NRAS, SF3B1 and EZH2. Diagnostics (Basel) 2021; 11:diagnostics11040600. [PMID: 33801781 PMCID: PMC8065453 DOI: 10.3390/diagnostics11040600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/16/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022] Open
Abstract
B-cell and T-cell lymphomas and leukemias often have distinct genetic mutations that are diagnostically defining or prognostically significant. A subset of these mutations consists of specific point mutations, which can be evaluated using genetic sequencing approaches or point mutation specific antibodies. Here, we describe genes harboring point mutations relevant to B-cell and T-cell malignancies and discuss the current availability of these targeted point mutation specific antibodies. We also evaluate the possibility of generating novel antibodies against known point mutations by computationally assessing for chemical and structural features as well as epitope antigenicity of these targets. Our results not only summarize several genetic mutations and identify existing point mutation specific antibodies relevant to hematologic malignancies, but also reveal potential underdeveloped targets which merit further study.
Collapse
|
11
|
Studying Sjögren's syndrome in mice: What is the best available model? J Oral Biol Craniofac Res 2021; 11:245-255. [PMID: 33665074 DOI: 10.1016/j.jobcr.2020.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/04/2020] [Accepted: 12/05/2020] [Indexed: 01/18/2023] Open
Abstract
Sjögren's syndrome (SS) is a common autoimmune disease characterized by lymphocytic infiltration and destruction of exocrine glands. The disease manifests primarily in the salivary and lacrimal glands, but other organs are also involved, leading to dry mouth, dry eyes, and other extra-glandular manifestations. Studying the disease in humans is entailed with many limitations and restrictions; therefore, the need for a proper mouse model is mandatory. SS mouse models are categorized, depending on the disease emergence into spontaneous or experimentally manipulated models. The usefulness of each mouse model varies depending on the SS features exhibited by that model; each SS model has advanced our understanding of the disease pathogenesis. In this review article, we list all the available murine models which have been used to study SS and we comment on the characteristics exhibited by each mouse model to assist scientists to select the appropriate model for their specific studies. We also recommend a murine strain that is the most relevant to the ideal SS model, based on our experience acquired during previous and current investigations.
Collapse
|
12
|
Sedlmeier G, Al‐Rawi V, Buchert J, Yserentant K, Rothley M, Steshina A, Gräßle S, Wu R, Hurrle T, Richer W, Decraene C, Thiele W, Utikal J, Abuillan W, Tanaka M, Herten D, Hill CS, Garvalov BK, Jung N, Bräse S, Sleeman JP. Id1 and Id3 Are Regulated Through Matrix‐Assisted Autocrine BMP Signaling and Represent Therapeutic Targets in Melanoma. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Georg Sedlmeier
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Vanessa Al‐Rawi
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Justyna Buchert
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Klaus Yserentant
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
- College of Medical and Dental Sciences & School of Chemistry University of Birmingham Birmingham UK
- Centre of Membrane Proteins and Receptors (COMPARE) Universities of Birmingham and Nottingham UK
| | - Melanie Rothley
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Anastasia Steshina
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Simone Gräßle
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
- Institute of Biological and Chemical Systems – Functional Molecular Systems (IBCS‐FMS) Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 D‐76344 Eggenstein‐Leopoldshafen Germany
| | - Ruo‐Lin Wu
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Thomas Hurrle
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
| | - Wilfrid Richer
- CNRS UMR144 Translational Research Department Institut Curie PSL Research University 26 rue d'Ulm Paris Cedex 05 75248 France
| | - Charles Decraene
- CNRS UMR144 Translational Research Department Institut Curie PSL Research University 26 rue d'Ulm Paris Cedex 05 75248 France
| | - Wilko Thiele
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Jochen Utikal
- Skin Cancer Unit German Cancer Research Center (DKFZ) Im Neuenheimer Feld 280 69120 Heidelberg Germany
- Department of Dermatology, Venereology and Allergology University Medical Center Mannheim Ruprecht‐Karl University of Heidelberg Theodor‐Kutzer‐Ufer 1–3 68167 Mannheim Germany
| | - Wasim Abuillan
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
| | - Motomu Tanaka
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
- Center for Integrative Medicine and Physics Institute for Advanced Study Kyoto University Yoshida Ushinomiya‐cho Sakyo‐Ku Kyoto 606‐8501 Japan
- Center for Integrative Medicine and Physics Institute for Advanced Study, Kyoto University Kyoto 606‐8501 Japan
| | - Dirk‐Peter Herten
- Institute of Physical Chemistry University of Heidelberg Im Neuenheimer Feld 229 69120 Heidelberg Germany
- College of Medical and Dental Sciences & School of Chemistry University of Birmingham Birmingham UK
- Centre of Membrane Proteins and Receptors (COMPARE) Universities of Birmingham and Nottingham UK
| | | | - Boyan K. Garvalov
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
| | - Nicole Jung
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
- Institute of Biological and Chemical Systems – Functional Molecular Systems (IBCS‐FMS) Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 D‐76344 Eggenstein‐Leopoldshafen Germany
| | - Stefan Bräse
- Institute of Organic Chemistry (IOC) Karlsruhe Institute of Technology Campus South, Building 30.42, Fritz‐Haber‐Weg 6 76131 Karlsruhe Germany
- Institute of Biological and Chemical Systems – Functional Molecular Systems (IBCS‐FMS) Karlsruhe Institute of Technology (KIT) Hermann‐von‐Helmholtz‐Platz 1 D‐76344 Eggenstein‐Leopoldshafen Germany
| | - Jonathan P. Sleeman
- European Center for Angioscience (ECAS) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Mannheim Institute for Innate Immunoscience (MI3) Medical Faculty Mannheim of the University of Heidelberg Ludolf‐Krehl‐Strasse 13–17 68167 Mannheim Germany
- Institute of Biological and Chemical Systems – Biological Information Processing (IBCS‐BIP) Karlsruhe Institute of Technology Campus North, Building 319, Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| |
Collapse
|
13
|
Misiou A, Garmey JC, Hensien JM, Harmon DB, Osinski V, McSkimming C, Marshall MA, Fischer JW, Grandoch M, McNamara CA. Helix-Loop-Helix Factor Id3 (Inhibitor of Differentiation 3): A Novel Regulator of Hyaluronan-Mediated Adipose Tissue Inflammation. Arterioscler Thromb Vasc Biol 2021; 41:796-807. [PMID: 33380173 PMCID: PMC8105274 DOI: 10.1161/atvbaha.120.315588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The aim of this study was to unravel mechanisms whereby deficiency of the transcription factor Id3 (inhibitor of differentiation 3) leads to metabolic dysfunction in visceral obesity. We investigated the impact of loss of Id3 on hyaluronic acid (HA) production by the 3 HAS isoenzymes (HA synthases; -1, -2, and -3) and on obesity-induced adipose tissue (AT) accumulation of proinflammatory B cells. Approach and Results: Male Id3-/- mice and respective wild-type littermate controls were fed a 60% high-fat diet for 4 weeks. An increase in inflammatory B2 cells was detected in Id3-/- epididymal AT. HA accumulated in epididymal AT of high-fat diet-fed Id3-/- mice and circulating levels of HA were elevated. Has2 mRNA expression was increased in epididymal AT of Id3-/- mice. Luciferase promoter assays showed that Id3 suppressed Has2 promoter activity, while loss of Id3 stimulated Has2 promoter activity. Functionally, HA strongly promoted B2 cell adhesion in the AT and on cultured vascular smooth muscle cells of Id3-/- mice, an effect sensitive to hyaluronidase. CONCLUSIONS Our data demonstrate that loss of Id3 increases Has2 expression in the epididymal AT, thereby promoting HA accumulation. In turn, elevated HA content promotes HA-dependent binding of B2 cells and an increase in the B2 cells in the AT, which contributes to AT inflammation.
Collapse
MESH Headings
- Adipose Tissue/immunology
- Adipose Tissue/metabolism
- Animals
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cell Adhesion
- Cells, Cultured
- Coculture Techniques
- Diet, High-Fat
- Disease Models, Animal
- Hyaluronan Synthases/genetics
- Hyaluronan Synthases/metabolism
- Hyaluronic Acid/biosynthesis
- Inhibitor of Differentiation Proteins/genetics
- Inhibitor of Differentiation Proteins/metabolism
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/immunology
- Myocytes, Smooth Muscle/metabolism
- Panniculitis/genetics
- Panniculitis/immunology
- Panniculitis/metabolism
- Phenotype
- Signal Transduction
- Up-Regulation
- Mice
Collapse
Affiliation(s)
- Angelina Misiou
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - James C. Garmey
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Jack M. Hensien
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Daniel B. Harmon
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Victoria Osinski
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Chantel McSkimming
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Melissa A. Marshall
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Jens W. Fischer
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Maria Grandoch
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Coleen A. McNamara
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Medicine, Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
14
|
Abstract
Somatic mutations are the driving force of many age-related diseases such as cancer and hematopoietic failure. A challenge in the field is to evaluate health impact of somatic mutations prior to the appearance of disease symptoms. We describe a genetic tool named MASCOT (mosaic analysis system with Cre or Tomato) for mosaic analysis of somatic mutations that drive clonal hematopoiesis and lymphomagenesis. MASCOT can be applied to mosaic analysis of broad tissue types, and thus provides a valuable tool to aid functional dissection of somatic mutations in studies of development and disease. Somatic mutations are major genetic contributors to cancers and many other age-related diseases. Many disease-causing somatic mutations can initiate clonal growth prior to the appearance of any disease symptoms, yet experimental models that can be used to examine clonal abnormalities are limited. We describe a mosaic analysis system with Cre or Tomato (MASCOT) for tracking mutant cells and demonstrate its utility for modeling clonal hematopoiesis. MASCOT can be induced to constitutively express either Cre-GFP or Tomato for lineage tracing of a mutant and a reference group of cells simultaneously. We conducted mosaic analysis to assess functions of the Id3 and/or Tet2 gene in hematopoietic cell development and clonal hematopoiesis. Using Tomato-positive cells as a reference population, we demonstrated the high sensitivity of this system for detecting cell-intrinsic phenotypes during short-term or long-term tracking of hematopoietic cells. Long-term tracking of Tet2 mutant or Tet2/Id3 double-mutant cells in our MASCOT model revealed a dynamic shift from myeloid expansion to lymphoid expansion and subsequent development of lymphoma. This work demonstrates the utility of the MASCOT method in mosaic analysis of single or combined mutations, making the system suitable for modeling somatic mutations identified in humans.
Collapse
|
15
|
Abstract
Epstein-Barr virus (EBV) infects human B cells and reprograms them to allow virus replication and persistence. One key viral factor in this process is latent membrane protein 2A (LMP2A), which has been described as a B cell receptor (BCR) mimic promoting malignant transformation. However, how LMP2A signaling contributes to tumorigenesis remains elusive. By comparing LMP2A and BCR signaling in primary human B cells using phosphoproteomics and transcriptome profiling, we identified molecular mechanisms through which LMP2A affects B cell biology. Consistent with the literature, we found that LMP2A mimics a subset of BCR signaling events, including tyrosine phosphorylation of the kinase SYK, the calcium initiation complex consisting of BLNK, BTK, and PLCγ2, and its downstream transcription factor NFAT. However, the majority of LMP2A-induced signaling events markedly differed from those induced by BCR stimulation. These included differential phosphorylation of kinases, phosphatases, adaptor proteins, transcription factors such as nuclear factor κB (NF-κB) and TCF3, as well as widespread changes in the transcriptional output of LMP2A-expressing B cells. LMP2A affected apoptosis and cell-cycle checkpoints by dysregulating the expression of apoptosis regulators such as BCl-xL and the tumor suppressor retinoblastoma-associated protein 1 (RB1). LMP2A cooperated with MYC and mutant cyclin D3, two oncogenic drivers of Burkitt lymphoma, to promote proliferation and survival of primary human B cells by counteracting MYC-induced apoptosis and by inhibiting RB1 function, thereby promoting cell-cycle progression. Our results indicate that LMP2A is not a pure BCR mimic but rather rewires intracellular signaling in EBV-infected B cells that optimizes cell survival and proliferation, setting the stage for oncogenic transformation.
Collapse
|
16
|
Gao Y, Chen Y, Zhang Z, Yu X, Zheng J. Recent Advances in Mouse Models of Sjögren's Syndrome. Front Immunol 2020; 11:1158. [PMID: 32695097 PMCID: PMC7338666 DOI: 10.3389/fimmu.2020.01158] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
Sjögren's syndrome (SS) is a complex rheumatoid disease that mainly affects exocrine glands, resulting in xerostomia (dry mouth) and xerophthalmia (dry eye). SS is characterized by autoantibodies, infiltration into exocrine glands, and ectopic expression of MHC II molecules on glandular epithelial cells. In contrast to the well-characterized clinical and immunological features, the etiology and pathogenesis of SS remain largely unknown. Animal models are powerful research tools for elucidating the pathogenesis of human diseases. To date, many mouse models of SS, including induced models, in which disease is induced in mice, and genetic models, in which mice spontaneously develop SS-like disease, have been established. These mouse models have provided new insight into the pathogenesis of SS. In this review, we aim to provide a comprehensive overview of recent advances in the field of experimental SS.
Collapse
Affiliation(s)
- Yunzhen Gao
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Yan Chen
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Zhongjian Zhang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Xinhua Yu
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), Borstel, Germany
| | - Junfeng Zheng
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
17
|
Hidalgo Y, Núñez S, Fuenzalida MJ, Flores-Santibáñez F, Sáez PJ, Dorner J, Lennon-Dumenil AM, Martínez V, Zorn E, Rosemblatt M, Sauma D, Bono MR. Thymic B Cells Promote Germinal Center-Like Structures and the Expansion of Follicular Helper T Cells in Lupus-Prone Mice. Front Immunol 2020; 11:696. [PMID: 32411134 PMCID: PMC7199236 DOI: 10.3389/fimmu.2020.00696] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/27/2020] [Indexed: 12/24/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the activation of autoreactive T and B cells, autoantibody production, and immune complex deposition in various organs. Previous evidence showed abnormal accumulation of B cells in the thymus of lupus-prone mice, but the role of this population in the progression of the disease remains mostly undefined. Here we analyzed the spatial distribution, function, and properties of this thymic B cell population in the BWF1 murine model of SLE. We found that in diseased animals, thymic B cells proliferate, and cluster in structures that resemble ectopic germinal centers. Moreover, we detected antibody-secreting cells in the thymus of diseased-BWF1 mice that produce anti-dsDNA IgG autoantibodies. We also found that thymic B cells from diseased-BWF1 mice induced the differentiation of thymocytes to follicular helper T cells (TFH). These data suggest that the accumulation of B cells in the thymus of BWF1 mice results in the formation of germinal center-like structures and the expansion of a TFH population, which may, in turn, activate and differentiate B cells into autoreactive plasma cells. Therefore, the thymus emerges as an important niche that supports the maintenance of the pathogenic humoral response in the development of murine SLE.
Collapse
Affiliation(s)
- Yessia Hidalgo
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Cells for Cells-Consorcio Regenero, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | | | - Maria Jose Fuenzalida
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Fundacion Ciencia & Vida, Santiago, Chile
| | | | - Pablo J Sáez
- INSERM U932, Institut Curie, Centre de Recherche, PSL Research University, Paris, France
| | - Jessica Dorner
- FAVET-INBIOGEN, Faculty of Veterinary Sciences, University of Chile, Santiago, Chile
| | | | - Victor Martínez
- FAVET-INBIOGEN, Faculty of Veterinary Sciences, University of Chile, Santiago, Chile
| | - Emmanuel Zorn
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| | - Mario Rosemblatt
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Fundacion Ciencia & Vida, Santiago, Chile.,Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Daniela Sauma
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Maria Rosa Bono
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
18
|
Gadomski S, Singh SK, Singh S, Sarkar T, Klarmann KD, Berenschot M, Seaman S, Jakubison B, Gudmundsson KO, Lockett S, Keller JR. Id1 and Id3 Maintain Steady-State Hematopoiesis by Promoting Sinusoidal Endothelial Cell Survival and Regeneration. Cell Rep 2020; 31:107572. [PMID: 32348770 PMCID: PMC8459380 DOI: 10.1016/j.celrep.2020.107572] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/19/2020] [Accepted: 04/02/2020] [Indexed: 02/08/2023] Open
Abstract
Investigating mechanisms that regulate endothelial cell (EC) growth and survival is important for understanding EC homeostasis and how ECs maintain stem cell niches. We report here that targeted loss of Id genes in adult ECs results in dilated, leaky sinusoids and a pro-inflammatory state that increases in severity over time. Disruption in sinusoidal integrity leads to increased hematopoietic stem cell (HSC) proliferation, differentiation, migration, and exhaustion. Mechanistically, sinusoidal ECs (SECs) show increased apoptosis because of reduced Bcl2-family gene expression following Id gene ablation. Furthermore, Id1-/-Id3-/- SECs and upstream type H vessels show increased expression of cyclin-dependent kinase inhibitors p21 and p27 and impaired ability to proliferate, which is rescued by reducing E2-2 expression. Id1-/-Id3-/- mice do not survive sublethal irradiation because of impaired vessel regeneration and hematopoietic failure. Thus, Id genes are required for the survival and regeneration of BM SECs during homeostasis and stress to maintain HSC development.
Collapse
Affiliation(s)
- Stephen Gadomski
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Satyendra K Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Shweta Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Tanmoy Sarkar
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Kimberly D Klarmann
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Maximillian Berenschot
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Steven Seaman
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA
| | - Brad Jakubison
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Kristbjorn O Gudmundsson
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Stephen Lockett
- Optical Microscopy and Analysis Laboratory, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jonathan R Keller
- Mouse Cancer Genetics Program, Center for Cancer Research, NCI, Frederick, MD 21702, USA; Basic Science Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA.
| |
Collapse
|
19
|
de Barrios O, Meler A, Parra M. MYC's Fine Line Between B Cell Development and Malignancy. Cells 2020; 9:E523. [PMID: 32102485 PMCID: PMC7072781 DOI: 10.3390/cells9020523] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
The transcription factor MYC is transiently expressed during B lymphocyte development, and its correct modulation is essential in defined developmental transitions. Although temporary downregulation of MYC is essential at specific points, basal levels of expression are maintained, and its protein levels are not completely silenced until the B cell becomes fully differentiated into a plasma cell or a memory B cell. MYC has been described as a proto-oncogene that is closely involved in many cancers, including leukemia and lymphoma. Aberrant expression of MYC protein in these hematological malignancies results in an uncontrolled rate of proliferation and, thereby, a blockade of the differentiation process. MYC is not activated by mutations in the coding sequence, and, as reviewed here, its overexpression in leukemia and lymphoma is mainly caused by gene amplification, chromosomal translocations, and aberrant regulation of its transcription. This review provides a thorough overview of the role of MYC in the developmental steps of B cells, and of how it performs its essential function in an oncogenic context, highlighting the importance of appropriate MYC regulation circuitry.
Collapse
Affiliation(s)
| | | | - Maribel Parra
- Lymphocyte Development and Disease Group, Josep Carreras Leukaemia Research Institute, IJC Building, Campus ICO-Germans Trias i Pujol, Ctra de Can Ruti, 08916 Barcelona, Spain (A.M.)
| |
Collapse
|
20
|
Avecilla V. Effect of Transcriptional Regulator ID3 on Pulmonary Arterial Hypertension and Hereditary Hemorrhagic Telangiectasia. Int J Vasc Med 2019; 2019:2123906. [PMID: 31380118 PMCID: PMC6657613 DOI: 10.1155/2019/2123906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/26/2019] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) can be discovered in patients who have a loss of function mutation of activin A receptor-like type 1 (ACVRL1) gene, a bone morphogenetic protein (BMP) type 1 receptor. Additionally, ACVRL1 mutations can lead to hereditary hemorrhagic telangiectasia (HHT), also known as Rendu-Osler-Weber disease, an autosomal dominant inherited disease that results in mucocutaneous telangiectasia and arteriovenous malformations (AVMs). Transcriptional regulator Inhibitor of DNA-Binding/Differentiation-3 (ID3) has been demonstrated to be involved in both PAH and HTT; however, the role of its overlapping molecular mechanistic effects has yet to be seen. This review will focus on the existing understanding of how ID3 may contribute to molecular involvement and perturbations thus altering both PAH and HHT outcomes. Improved understanding of how ID3 mediates these pathways will likely provide knowledge in the inhibition and regulation of these diseases through targeted therapies.
Collapse
Affiliation(s)
- Vincent Avecilla
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA
- Celgene Corporation, Summit, NJ 07901, USA
| |
Collapse
|
21
|
Vivino FB, Bunya VY, Massaro-Giordano G, Johr CR, Giattino SL, Schorpion A, Shafer B, Peck A, Sivils K, Rasmussen A, Chiorini JA, He J, Ambrus JL. Sjogren's syndrome: An update on disease pathogenesis, clinical manifestations and treatment. Clin Immunol 2019; 203:81-121. [PMID: 31022578 DOI: 10.1016/j.clim.2019.04.009] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 04/19/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Frederick B Vivino
- Penn Sjögren's Center, Penn Presbyterian Medical Center, University of Pennsylvania Perelman School of Medicine, 3737 Market Street, Philadelphia, PA 19104, USA.
| | - Vatinee Y Bunya
- Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, 51 N. 39(th) Street, Philadelphia, PA 19104, USA.
| | - Giacomina Massaro-Giordano
- Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, 51 N. 39(th) Street, Philadelphia, PA 19104, USA.
| | - Chadwick R Johr
- Penn Sjögren's Center, Penn Presbyterian Medical Center, University of Pennsylvania Perelman School of Medicine, 3737 Market Street, Philadelphia, PA 19104, USA.
| | - Stephanie L Giattino
- Penn Sjögren's Center, Penn Presbyterian Medical Center, University of Pennsylvania Perelman School of Medicine, 3737 Market Street, Philadelphia, PA 19104, USA.
| | - Annemarie Schorpion
- Penn Sjögren's Center, Penn Presbyterian Medical Center, University of Pennsylvania Perelman School of Medicine, 3737 Market Street, Philadelphia, PA 19104, USA.
| | - Brian Shafer
- Scheie Eye Institute, University of Pennsylvania Perelman School of Medicine, 51 N. 39(th) Street, Philadelphia, PA 19104, USA.
| | - Ammon Peck
- Department of Infectious Diseases and Immunology, University of Florida College of Veterinary Medicine, PO Box 100125, Gainesville, FL 32610, USA.
| | - Kathy Sivils
- Oklahoma Medical Research Foundation, Arthritis and Clinical Immunology Program, 825 NE 13th Street, OK 73104, USA.
| | - Astrid Rasmussen
- Oklahoma Medical Research Foundation, Arthritis and Clinical Immunology Program, 825 NE 13th Street, OK 73104, USA.
| | - John A Chiorini
- NIH, Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, Building 10, Room 1n113, 10 Center DR Msc 1190, Bethesda, MD 20892-1190, USA.
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing 100044, China
| | - Julian L Ambrus
- Division of Allergy, Immunology and Rheumatology, SUNY at Buffalo School of Medicine, 100 High Street, Buffalo, NY 14203, USA.
| |
Collapse
|
22
|
Amaral LHP, Bufalo NE, Peres KC, Barreto IS, Campos AHJFM, Ward LS. ID Proteins May Reduce Aggressiveness of Thyroid Tumors. Endocr Pathol 2019; 30:24-30. [PMID: 30413933 DOI: 10.1007/s12022-018-9556-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
ID genes have an important function in the cell cycle, and ID proteins may help identify aggressive tumors, besides being considered promising therapeutic targets. However, their role in thyroid tumors is still poorly understood. We examined ID expression and their correlation with diagnostic and prognostic features aiming to find a clinical application in differentiated thyroid carcinoma (DTC) cases. mRNA levels of ID1, ID2, ID3, and ID4 genes were quantified and their expression was observed by immunohistochemistry in 194 thyroid samples including 68 goiters, 16 follicular adenomas, 75 classic papillary thyroid carcinomas, 18 follicular variants of papillary thyroid carcinoma, 5 follicular thyroid carcinomas, and 1 anaplastic thyroid cancer, besides 11 normal thyroid tissues. DTC patients were managed according to standard protocols and followed up for M = 28 ± 16 months. ID2, ID3, and ID4 mRNA levels were higher in benign (2.0 ± 1.9; 0.6 ± 0.6; and 0.7 ± 1.0 AU, respectively) than those in malignant nodules (0.30 ± 0.62; 0.3 ± 0.3; and 0.2 ± 0.3 AU, respectively, p < 0.0001 for all three genes) and were associated with no extra thyroid invasion or metastasis at diagnosis. ID3 nuclear protein expression was higher in benign than that in malignant cells (5.2 ± 0.9 vs 3.0 ± 1.8 AU; p < 0.0001). On the contrary, the cytoplasmic expression of ID3 was higher in malignant than that in benign lesions (5.7 ± 1.5 vs 4.0 ± 1.4 AU; p < 0.0001). Our data indicate that ID genes are involved in thyroid tumorigenesis and suggest these genes act impeding the evolution of more aggressive phenotypes. The different patterns of their tissue expression may help identify malignancy and characterize thyroid lesion aggressiveness.
Collapse
Affiliation(s)
- Laís Helena Pereira Amaral
- Laboratory of Cancer Molecular Genetics, School of Medical Sciences, State University of Campinas (Unicamp), Campinas, SP, Brazil.
- , Pouso Alegre, Brazil.
| | - Natássia Elena Bufalo
- Laboratory of Cancer Molecular Genetics, School of Medical Sciences, State University of Campinas (Unicamp), Campinas, SP, Brazil
| | - Karina Colombera Peres
- Laboratory of Cancer Molecular Genetics, School of Medical Sciences, State University of Campinas (Unicamp), Campinas, SP, Brazil
| | - Icléia Siqueira Barreto
- Department of Pathology, School of Medical Sciences, State University of Campinas (Unicamp), Campinas, SP, Brazil
| | | | - Laura Sterian Ward
- Laboratory of Cancer Molecular Genetics, School of Medical Sciences, State University of Campinas (Unicamp), Campinas, SP, Brazil
| |
Collapse
|
23
|
Doke M, Das J, Felty Q. Letter to the Editor: Is Id3 proliferative or antiproliferative? Am J Physiol Lung Cell Mol Physiol 2018; 315:L334-L335. [PMID: 30088801 DOI: 10.1152/ajplung.00205.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Mayur Doke
- Department of Environmental Health Sciences, Florida International University , Miami, Florida
| | - Jayanta Das
- Department of Environmental Health Sciences, Florida International University , Miami, Florida
| | - Quentin Felty
- Department of Environmental Health Sciences, Florida International University , Miami, Florida
| |
Collapse
|
24
|
Zhou H, Wang L, Xu Q, Liu Q, Liu H, Qiu W, Hu T, Lv Y, Zhang Q. ID3 may protect mice from anti‑GBM glomerulonephritis by regulating the differentiation of Th17 and Treg cells. Mol Med Rep 2017; 16:9086-9094. [PMID: 28990057 DOI: 10.3892/mmr.2017.7724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 07/31/2017] [Indexed: 11/05/2022] Open
Abstract
Anti‑glomerular basement membrane glomerulonephritis (anti‑GBM GN) is an autoimmune disease that leads to severe and rapidly progressive renal injury. Inhibition of DNA‑binding factor 3 (ID3) serves a key role in autoimmune diseases, such as asthma and Sjögren's syndrome, and in experimental allergic encephalitis models. However, the role of ID3 in the progression of anti‑GBM GN remains unknown. In the present study, ID3 mRNA expression increased between 3‑ and 20‑fold in the renal tissues of anti‑GBM GN mice compared with the Control group, with a peak at day 14 post‑induction. In addition, ID3 protein expression was upregulated from day 7 onwards. The expression of ID3 was also examined in the spleen, and was demonstrated to be increased in the spleen of nephritic mice. T helper 17 (Th17) cells and regulatory T (Treg) cells were present throughout the entire period of observation (from day 7 to day 28) in anti‑GBM GN mice, which may vary at different time points, accompanied with the expression of ID3. In vitro, ID3 expression was increased when CD4+ T cells differentiated into Tregs; however, expression was lower in Th17 cells. Following treatment with ID3 small interfering RNA, RAR‑related orphan receptor γt, but not forkhead box P3, expression increased. Furthermore, increased expression of interleukin‑17A was also observed when ID3 was blocked. In addition, ID3 was able to interact with transcription factor E2A. A significant increase in binding between ID3 and E2A was observed in anti‑GBM GN from day 7 onwards, with a peak at day 14 in both renal tissue and spleen. In conclusion, ID3 may be involved in the differentiation of Th17 and Tregs by downregulating Th17 cells, which is probably associated with binding to E2A. The present results suggested that ID3 may offer protection against anti‑GBM GN in mice.
Collapse
Affiliation(s)
- Huan Zhou
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Le Wang
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qing Xu
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qingquan Liu
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Hui Liu
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Wenhui Qiu
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Tingyang Hu
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yongman Lv
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qian Zhang
- Division of Nephrology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
25
|
Behdani E, Bakhtiarizadeh MR. Construction of an integrated gene regulatory network link to stress-related immune system in cattle. Genetica 2017; 145:441-454. [PMID: 28825201 DOI: 10.1007/s10709-017-9980-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 08/14/2017] [Indexed: 01/01/2023]
Abstract
The immune system is an important biological system that is negatively impacted by stress. This study constructed an integrated regulatory network to enhance our understanding of the regulatory gene network used in the stress-related immune system. Module inference was used to construct modules of co-expressed genes with bovine leukocyte RNA-Seq data. Transcription factors (TFs) were then assigned to these modules using Lemon-Tree algorithms. In addition, the TFs assigned to each module were confirmed using the promoter analysis and protein-protein interactions data. Therefore, our integrated method identified three TFs which include one TF that is previously known to be involved in immune response (MYBL2) and two TFs (E2F8 and FOXS1) that had not been recognized previously and were identified for the first time in this study as novel regulatory candidates in immune response. This study provides valuable insights on the regulatory programs of genes involved in the stress-related immune system.
Collapse
Affiliation(s)
- Elham Behdani
- Department of Animal Sciences, College of Agriculture and Natural Resources, Ramin University, Khozestan, Iran
| | | |
Collapse
|
26
|
Contribution of Inhibitor of DNA Binding/Differentiation-3 and Endocrine Disrupting Chemicals to Pathophysiological Aspects of Chronic Disease. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6307109. [PMID: 28785583 PMCID: PMC5530454 DOI: 10.1155/2017/6307109] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/15/2017] [Accepted: 05/29/2017] [Indexed: 12/12/2022]
Abstract
The overwhelming increase in the global incidence of obesity and its associated complications such as insulin resistance, atherosclerosis, pulmonary disease, and degenerative disorders including dementia constitutes a serious public health problem. The Inhibitor of DNA Binding/Differentiation-3 (ID3), a member of the ID family of transcriptional regulators, has been shown to play a role in adipogenesis and therefore ID3 may influence obesity and metabolic health in response to environmental factors. This review will highlight the current understanding of how ID3 may contribute to complex chronic diseases via metabolic perturbations. Based on the increasing number of reports that suggest chronic exposure to and accumulation of endocrine disrupting chemicals (EDCs) within the human body are associated with metabolic disorders, we will also consider the impact of these chemicals on ID3. Improved understanding of the ID3 pathways by which exposure to EDCs can potentiate complex chronic diseases in populations with metabolic disorders (obesity, metabolic syndrome, and glucose intolerance) will likely provide useful knowledge in the prevention and control of complex chronic diseases associated with exposure to environmental pollutants.
Collapse
|
27
|
Abstract
Inhibitors of DNA binding and cell differentiation (Id) proteins are members of the large family of the helix-loop-helix (HLH) transcription factors, but they lack any DNA-binding motif. During development, the Id proteins play a key role in the regulation of cell-cycle progression and cell differentiation by modulating different cell-cycle regulators both by direct and indirect mechanisms. Several Id-protein interacting partners have been identified thus far, which belong to structurally and functionally unrelated families, including, among others, the class I and II bHLH transcription factors, the retinoblastoma protein and related pocket proteins, the paired-box transcription factors, and the S5a subunit of the 26 S proteasome. Although the HLH domain of the Id proteins is involved in most of their protein-protein interaction events, additional motifs located in their N-terminal and C-terminal regions are required for the recognition of diverse protein partners. The ability of the Id proteins to interact with structurally different proteins is likely to arise from their conformational flexibility: indeed, these proteins contain intrinsically disordered regions that, in the case of the HLH region, undergo folding upon self- or heteroassociation. Besides their crucial role for cell-fate determination and cell-cycle progression during development, other important cellular events have been related to the Id-protein expression in a number of pathologies. Dysregulated Id-protein expression has been associated with tumor growth, vascularization, invasiveness, metastasis, chemoresistance and stemness, as well as with various developmental defects and diseases. Herein we provide an overview on the structural properties, mode of action, biological function and therapeutic potential of these regulatory proteins.
Collapse
Affiliation(s)
- Cornelia Roschger
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria
| | - Chiara Cabrele
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria.
| |
Collapse
|
28
|
Roschger C, Cabrele C. The Id-protein family in developmental and cancer-associated pathways. Cell Commun Signal 2017; 15:7. [PMID: 28122577 PMCID: PMC5267474 DOI: 10.1186/s12964-016-0161-y] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/29/2016] [Indexed: 01/15/2023] Open
Abstract
Inhibitors of DNA binding and cell differentiation (Id) proteins are members of the large family of the helix-loop-helix (HLH) transcription factors, but they lack any DNA-binding motif. During development, the Id proteins play a key role in the regulation of cell-cycle progression and cell differentiation by modulating different cell-cycle regulators both by direct and indirect mechanisms. Several Id-protein interacting partners have been identified thus far, which belong to structurally and functionally unrelated families, including, among others, the class I and II bHLH transcription factors, the retinoblastoma protein and related pocket proteins, the paired-box transcription factors, and the S5a subunit of the 26 S proteasome. Although the HLH domain of the Id proteins is involved in most of their protein-protein interaction events, additional motifs located in their N-terminal and C-terminal regions are required for the recognition of diverse protein partners. The ability of the Id proteins to interact with structurally different proteins is likely to arise from their conformational flexibility: indeed, these proteins contain intrinsically disordered regions that, in the case of the HLH region, undergo folding upon self- or heteroassociation. Besides their crucial role for cell-fate determination and cell-cycle progression during development, other important cellular events have been related to the Id-protein expression in a number of pathologies. Dysregulated Id-protein expression has been associated with tumor growth, vascularization, invasiveness, metastasis, chemoresistance and stemness, as well as with various developmental defects and diseases. Herein we provide an overview on the structural properties, mode of action, biological function and therapeutic potential of these regulatory proteins.
Collapse
Affiliation(s)
- Cornelia Roschger
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria
| | - Chiara Cabrele
- Department of Molecular Biology, University of Salzburg, Billrothstrasse 11, Salzburg, 5020, Austria.
| |
Collapse
|
29
|
Id3 Orchestrates Germinal Center B Cell Development. Mol Cell Biol 2016; 36:2543-52. [PMID: 27457619 DOI: 10.1128/mcb.00150-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 07/18/2016] [Indexed: 11/20/2022] Open
Abstract
Previous studies have demonstrated that E proteins induce activation-induced deaminase (AID) expression in activated B cells. Here, we examined the role of Id3 in germinal center (GC) cells. We found that Id3 expression is high in follicular B lineage cells but declines in GC cells. Immunized mice with Id3 expression depleted displayed a block in germinal center B cell maturation, showed reduced numbers of marginal zone B cells and class-switched cells, and were associated with decreased antibody titers and lower numbers of plasma cells. In vitro, Id3-depleted B cells displayed a defect in class switch recombination. Whereas AID levels were not altered in Id3-depleted activated B cells, the expression of a subset of genes encoding signaling components of antigen receptor-, cytokine receptor-, and chemokine receptor-mediated signaling was significantly impaired. We propose that during the GC reaction, Id3 levels decline to activate the expression of genes encoding signaling components that mediate B cell receptor- and or cytokine receptor-mediated signaling to promote the differentiation of GC B cells.
Collapse
|
30
|
Selesniemi K, Albers RE, Brown TL. Id2 Mediates Differentiation of Labyrinthine Placental Progenitor Cell Line, SM10. Stem Cells Dev 2016; 25:959-74. [PMID: 27168216 PMCID: PMC4931356 DOI: 10.1089/scd.2016.0010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/17/2016] [Indexed: 11/12/2022] Open
Abstract
The placenta is an organ that is formed transiently during pregnancy, and appropriate placental development is necessary for fetal survival and growth. Proper differentiation of the labyrinthine layer of the placenta is especially crucial, as it establishes the fetal-maternal interface that is involved in physiological exchange processes. Although previous studies have indicated the importance of inhibitor of differentiation/inhibitor of DNA binding-2 (Id2) helix-loop-helix transcriptional regulator in mediating cell differentiation, the ability of Id2 to regulate differentiation toward the labyrinthine (transport) lineage of the placenta has yet to be determined. In the current study, we have generated labyrinthine trophoblast progenitor cells with increased (SM10-Id2) or decreased (SM10-Id2-shRNA) Id2 expression and determined the effect on TGF-β-induced differentiation. Our Id2 overexpression and knockdown analyses indicate that Id2 mediates TGF-β-induced morphological differentiation of labyrinthine trophoblast cells, as Id2 overexpression prevents differentiation and Id2 knockdown results in differentiation. Thus, our data indicate that Id2 is an important molecular mediator of labyrinthine trophoblast differentiation. An understanding of the regulators of trophoblast progenitor differentiation toward the labyrinthine lineage may offer insights into events governing pregnancy-associated disorders, such as placental insufficiency, fetal growth restriction, and preeclampsia.
Collapse
Affiliation(s)
- Kaisa Selesniemi
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University Boonshoft School of Medicine , Dayton, Ohio
| | - Renee E Albers
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University Boonshoft School of Medicine , Dayton, Ohio
| | - Thomas L Brown
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University Boonshoft School of Medicine , Dayton, Ohio
| |
Collapse
|
31
|
Gloury R, Zotos D, Zuidscherwoude M, Masson F, Liao Y, Hasbold J, Corcoran LM, Hodgkin PD, Belz GT, Shi W, Nutt SL, Tarlinton DM, Kallies A. Dynamic changes in Id3 and E-protein activity orchestrate germinal center and plasma cell development. J Exp Med 2016; 213:1095-111. [PMID: 27217539 PMCID: PMC4886367 DOI: 10.1084/jem.20152003] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/15/2016] [Indexed: 12/27/2022] Open
Abstract
Gloury et al. reveal an essential role for the Id3–E-protein axis in the transcriptional regulation of humoral immunity. The generation of high-affinity antibodies requires germinal center (GC) development and differentiation of long-lived plasma cells in a multilayered process that is tightly controlled by the activity of multiple transcription factors. Here, we reveal a new layer of complexity by demonstrating that dynamic changes in Id3 and E-protein activity govern both GC and plasma cell differentiation. We show that down-regulation of Id3 in B cells is essential for releasing E2A and E2-2, which in a redundant manner are required for antigen-induced B cell differentiation. We demonstrate that this pathway controls the expression of multiple key factors, including Blimp1, Xbp1, and CXCR4, and is therefore critical for establishing the transcriptional network that controls GC B cell and plasma cell differentiation.
Collapse
Affiliation(s)
- Renee Gloury
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Dimitra Zotos
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Malou Zuidscherwoude
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Frederick Masson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yang Liao
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jhaguaral Hasbold
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Lynn M Corcoran
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Phil D Hodgkin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Gabrielle T Belz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Wei Shi
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Computing and Information Systems, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stephen L Nutt
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - David M Tarlinton
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Axel Kallies
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
32
|
Park YS, Gauna AE, Cha S. Mouse Models of Primary Sjogren's Syndrome. Curr Pharm Des 2016; 21:2350-64. [PMID: 25777752 DOI: 10.2174/1381612821666150316120024] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/13/2015] [Indexed: 01/03/2023]
Abstract
Sjogren's syndrome (SjS) is a chronic autoimmune disorder characterized by immune cell infiltration and progressive injury to the salivary and lacrimal glands. As a consequence, patients with SjS develop xerostomia (dry mouth) and keratoconjunctivitis sicca (dry eyes). SjS is the third most common rheumatic autoimmune disorder, affecting 4 million Americans with over 90% of patients being female. Current diagnostic criteria for SjS frequently utilize histological examinations of minor salivary glands for immune cell foci, serology for autoantibodies, and dry eye evaluation by corneal or conjunctival staining. SjS can be classified as primary or secondary SjS, depending on whether it occurs alone or in association with other systemic rheumatic conditions, respectively. Clinical manifestations typically become apparent when the disease is relatively advanced in SjS patients, which poses a challenge for early diagnosis and treatment of SjS. Therefore, SjS mouse models, because of their close resemblance to the human SjS, have been extremely valuable to identify early disease markers and to investigate underlying biological and immunological dysregulations. However, it is important to bear in mind that no single mouse model has duplicated all aspects of SjS pathogenesis and clinical features, mainly due to the multifactorial etiology of SjS that includes numerous susceptibility genes and environmental factors. As such, various mouse models have been developed in the field to try to recapitulate SjS. In this review, we focus on recent mouse models of primary SjS xerostomia and describe them under three categories of spontaneous, genetically engineered, and experimentally induced models. In addition, we discuss future perspectives highlighting pros and cons of utilizing mouse models and current demands for improved models.
Collapse
Affiliation(s)
| | | | - Seunghee Cha
- Department of Oral and Maxillofacial Diagnostic Sciences, University of Florida College of Dentistry, Gainesville, FL32610, USA.
| |
Collapse
|
33
|
Combined Id1 and Id3 Deletion Leads to Severe Erythropoietic Disturbances. PLoS One 2016; 11:e0154480. [PMID: 27128622 PMCID: PMC4851361 DOI: 10.1371/journal.pone.0154480] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 04/13/2016] [Indexed: 12/31/2022] Open
Abstract
The Inhibitor of DNA Binding (Id) proteins play a crucial role in regulating hematopoiesis and are known to interact with E proteins and the bHLH family of transcription factors. Current efforts seek to elucidate the individual roles of Id members in regulating hematopoietic development and specification. However, the nature of their functional redundancies remains elusive since ablation of multiple Id genes is embryonically lethal. We developed a model to test this compensation in the adult. We report that global Id3 ablation with Tie2Cre-mediated conditional ablation of Id1 in both hematopoietic and endothelial cells (Id cDKO) extends viability to 1 year but leads to multi-lineage hematopoietic defects including the emergence of anemia associated with defective erythroid development, a novel phenotype unreported in prior single Id knockout studies. We observe decreased cell counts in the bone marrow and splenomegaly to dimensions beyond what is seen in single Id knockout models. Transcriptional dysregulation of hematopoietic regulators observed in bone marrow cells is also magnified in the spleen. E47 protein levels were elevated in Id cDKO bone marrow cell isolates, but decreased in the erythroid lineage. Chromatin immunoprecipitation (ChIP) studies reveal increased occupancy of E47 and GATA1 at the promoter regions of β-globin and E2A. Bone marrow transplantation studies highlight the importance of intrinsic Id signals in maintaining hematopoietic homeostasis while revealing a strong extrinsic influence in the development of anemia. Together, these findings demonstrate that loss of Id compensation leads to dysregulation of the hematopoietic transcriptional network and multiple defects in erythropoietic development in adult mice.
Collapse
|
34
|
Harmon DB, Srikakulapu P, Kaplan JL, Oldham SN, McSkimming C, Garmey JC, Perry HM, Kirby JL, Prohaska TA, Gonen A, Hallowell P, Schirmer B, Tsimikas S, Taylor AM, Witztum JL, McNamara CA. Protective Role for B-1b B Cells and IgM in Obesity-Associated Inflammation, Glucose Intolerance, and Insulin Resistance. Arterioscler Thromb Vasc Biol 2016; 36:682-91. [PMID: 26868208 DOI: 10.1161/atvbaha.116.307166] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 02/01/2016] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Little is known about the role(s) B cells play in obesity-induced metabolic dysfunction. This study used a mouse with B-cell-specific deletion of Id3 (Id3(Bcell KO)) to identify B-cell functions involved in the metabolic consequences of obesity. APPROACH AND RESULTS Diet-induced obese Id3(Bcell KO) mice demonstrated attenuated inflammation and insulin resistance in visceral adipose tissue (VAT), and improved systemic glucose tolerance. VAT in Id3(Bcell KO) mice had increased B-1b B cells and elevated IgM natural antibodies to oxidation-specific epitopes. B-1b B cells reduced cytokine production in VAT M1 macrophages, and adoptively transferred B-1b B cells trafficked to VAT and produced natural antibodies for the duration of 13-week studies. B-1b B cells null for Id3 demonstrated increased proliferation, established larger populations in Rag1(-/-) VAT, and attenuated diet-induced glucose intolerance and VAT insulin resistance in Rag1(-/-) hosts. However, transfer of B-1b B cells unable to secrete IgM had no effect on glucose tolerance. In an obese human population, results provided the first evidence that B-1 cells are enriched in human VAT and IgM antibodies to oxidation-specific epitopes inversely correlated with inflammation and insulin resistance. CONCLUSIONS NAb-producing B-1b B cells are increased in Id3(Bcell KO) mice and attenuate adipose tissue inflammation and glucose intolerance in diet-induced obese mice. Additional findings are the first to identify VAT as a reservoir for human B-1 cells and to link anti-inflammatory IgM antibodies with reduced inflammation and improved metabolic phenotype in obese humans.
Collapse
Affiliation(s)
- Daniel B Harmon
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Prasad Srikakulapu
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Jennifer L Kaplan
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Stephanie N Oldham
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Chantel McSkimming
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - James C Garmey
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Heather M Perry
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Jennifer L Kirby
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Thomas A Prohaska
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Ayelet Gonen
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Peter Hallowell
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Bruce Schirmer
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Sotirios Tsimikas
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Angela M Taylor
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Joseph L Witztum
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.)
| | - Coleen A McNamara
- From the Cardiovascular Research Center (D.B.H., P.S., J.L.K., S.N.O., C.M.S., J.C.G., H.M.P., C.A.M.), Department of Biochemistry, Molecular Biology, and Genetics (D.B.H.), Division of Cardiovascular Medicine, Department of Medicine (P.S., A.M.T., C.A.M.), Department of Pathology (J.L.K., H.M.P.), Division of Endocrinology and Metabolism, Department of Medicine (J.L.K.), Department of Surgery (P.H., B.S.), Beirne B. Carter Center for Immunology Research (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, University of California San Diego, La Jolla (T.A.P., A.G., S.T., J.L.W.).
| |
Collapse
|
35
|
Jones SA, Toh AEJ, Odobasic D, Oudin MAV, Cheng Q, Lee JPW, White SJ, Russ BE, Infantino S, Light A, Tarlinton DM, Harris J, Morand EF. Glucocorticoid-induced leucine zipper (GILZ) inhibits B cell activation in systemic lupus erythematosus. Ann Rheum Dis 2015; 75:739-47. [PMID: 26612340 DOI: 10.1136/annrheumdis-2015-207744] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 11/01/2015] [Indexed: 01/29/2023]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) is a serious multisystem autoimmune disease, mediated by disrupted B cell quiescence and typically treated with glucocorticoids. We studied whether B cells in SLE are regulated by the glucocorticoid-induced leucine zipper (GILZ) protein, an endogenous mediator of anti-inflammatory effects of glucocorticoids. METHODS We conducted a study of GILZ expression in blood mononuclear cells of patients with SLE, performed in vitro analyses of GILZ function in mouse and human B cells, assessed the contributions of GILZ to autoimmunity in mice, and used the nitrophenol coupled to keyhole limpet haemocyanin model of immunisation in mice. RESULTS Reduced B cell GILZ was observed in patients with SLE and lupus-prone mice, and impaired induction of GILZ in patients with SLE receiving glucocorticoids was associated with increased disease activity. GILZ was downregulated in naïve B cells upon stimulation in vitro and in germinal centre B cells, which contained less enrichment of H3K4me3 at the GILZ promoter compared with naïve and memory B cells. Mice lacking GILZ spontaneously developed lupus-like autoimmunity, and GILZ deficiency resulted in excessive B cell responses to T-dependent stimulation. Accordingly, loss of GILZ in naïve B cells allowed upregulation of multiple genes that promote the germinal centre B cell phenotype, including lupus susceptibility genes and genes involved in cell survival and proliferation. Finally, treatment of human B cells with a cell-permeable GILZ fusion protein potently suppressed their responsiveness to T-dependent stimuli. CONCLUSIONS Our findings demonstrated that GILZ is a non-redundant regulator of B cell activity, with important potential clinical implications in SLE.
Collapse
Affiliation(s)
- Sarah A Jones
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia
| | - Andrew E J Toh
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia
| | - Dragana Odobasic
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia
| | - Marie-Anne Virginie Oudin
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia
| | - Qiang Cheng
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia
| | - Jacinta P W Lee
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia
| | - Stefan J White
- Department of Human Genetics, Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Brendan E Russ
- Department of Microbiology and Immunology, The Doherty Institute at The University of Melbourne, Parkville, Victoria, Australia
| | - Simona Infantino
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia Department of Experimental Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Amanda Light
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia Department of Experimental Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - David M Tarlinton
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia Department of Experimental Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - James Harris
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia
| | - Eric F Morand
- Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Clayton, Melbourne, Australia
| |
Collapse
|
36
|
Kramer JM, Holodick NE, Vizconde TC, Raman I, Yan M, Li QZ, Gaile DP, Rothstein TL. Analysis of IgM antibody production and repertoire in a mouse model of Sjögren's syndrome. J Leukoc Biol 2015; 99:321-31. [PMID: 26382297 DOI: 10.1189/jlb.2a0715-297r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/31/2015] [Indexed: 12/20/2022] Open
Abstract
This study tested the hypothesis that B cells from salivary tissue are distinct in terms of proliferative capacity, immunoglobulin M secretion, repertoire, and autoantibody enrichment in Sjögren's syndrome. We sorted purified B cells from the spleen, cervical lymph nodes, and submandibular glands of a primary Sjögren's syndrome mouse model (Id3(-/-)). Enzyme-linked immunospot and proliferation assays were performed with stimulated B cells. We single-cell sorted B cells from the spleen, cervical lymph nodes, and submandibular gland tissue from Sjögren's syndrome mice and sequenced immunoglobulin M heavy-chain variable regions. Finally, autoantigen arrays were performed using immunoglobulin M derived from sera, cervical lymph nodes, spleens, and submandibular gland tissue of Id3(-/-) animals. Results suggest B cells from salivary tissue of Sjögren's syndrome mice are similar to those from secondary immune sites in terms of proliferative and secretory capacity. However, differences in repertoire usage, heavy chain complementarity-determining region 3 length, mutational frequency, and N region addition were observed among B cells derived from submandibular gland, cervical lymph node, and spleen tissue. Moreover, autoantigen array data show immunoglobulin M from salivary B cells have enriched specificity for Ro (Sjögren's syndrome A) and La (Sjögren's syndrome B). All together, these data suggest salivary B cells have unique repertoire characteristics that likely influence autoantigen binding and contribute to Sjögren's syndrome disease in a tissue-specific manner.
Collapse
Affiliation(s)
- Jill M Kramer
- *Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, New York, USA; Division of Oral and Maxillofacial Pathology, Department of Dental Medicine, Long Island Jewish Medical Center, New Hyde Park, New York, USA; Department of Oral Biology, School of Dental Medicine, and Department of Biostatistics, State University of New York at Buffalo, Buffalo, New York, USA; Department of Dental Medicine and Medicine, Hofstra North Shore-LIJ School of Medicine, Hempstead, New York, USA; and Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nichol E Holodick
- *Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, New York, USA; Division of Oral and Maxillofacial Pathology, Department of Dental Medicine, Long Island Jewish Medical Center, New Hyde Park, New York, USA; Department of Oral Biology, School of Dental Medicine, and Department of Biostatistics, State University of New York at Buffalo, Buffalo, New York, USA; Department of Dental Medicine and Medicine, Hofstra North Shore-LIJ School of Medicine, Hempstead, New York, USA; and Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Teresa C Vizconde
- *Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, New York, USA; Division of Oral and Maxillofacial Pathology, Department of Dental Medicine, Long Island Jewish Medical Center, New Hyde Park, New York, USA; Department of Oral Biology, School of Dental Medicine, and Department of Biostatistics, State University of New York at Buffalo, Buffalo, New York, USA; Department of Dental Medicine and Medicine, Hofstra North Shore-LIJ School of Medicine, Hempstead, New York, USA; and Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Indu Raman
- *Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, New York, USA; Division of Oral and Maxillofacial Pathology, Department of Dental Medicine, Long Island Jewish Medical Center, New Hyde Park, New York, USA; Department of Oral Biology, School of Dental Medicine, and Department of Biostatistics, State University of New York at Buffalo, Buffalo, New York, USA; Department of Dental Medicine and Medicine, Hofstra North Shore-LIJ School of Medicine, Hempstead, New York, USA; and Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mei Yan
- *Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, New York, USA; Division of Oral and Maxillofacial Pathology, Department of Dental Medicine, Long Island Jewish Medical Center, New Hyde Park, New York, USA; Department of Oral Biology, School of Dental Medicine, and Department of Biostatistics, State University of New York at Buffalo, Buffalo, New York, USA; Department of Dental Medicine and Medicine, Hofstra North Shore-LIJ School of Medicine, Hempstead, New York, USA; and Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Quan-Zhen Li
- *Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, New York, USA; Division of Oral and Maxillofacial Pathology, Department of Dental Medicine, Long Island Jewish Medical Center, New Hyde Park, New York, USA; Department of Oral Biology, School of Dental Medicine, and Department of Biostatistics, State University of New York at Buffalo, Buffalo, New York, USA; Department of Dental Medicine and Medicine, Hofstra North Shore-LIJ School of Medicine, Hempstead, New York, USA; and Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel P Gaile
- *Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, New York, USA; Division of Oral and Maxillofacial Pathology, Department of Dental Medicine, Long Island Jewish Medical Center, New Hyde Park, New York, USA; Department of Oral Biology, School of Dental Medicine, and Department of Biostatistics, State University of New York at Buffalo, Buffalo, New York, USA; Department of Dental Medicine and Medicine, Hofstra North Shore-LIJ School of Medicine, Hempstead, New York, USA; and Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas L Rothstein
- *Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, New York, USA; Division of Oral and Maxillofacial Pathology, Department of Dental Medicine, Long Island Jewish Medical Center, New Hyde Park, New York, USA; Department of Oral Biology, School of Dental Medicine, and Department of Biostatistics, State University of New York at Buffalo, Buffalo, New York, USA; Department of Dental Medicine and Medicine, Hofstra North Shore-LIJ School of Medicine, Hempstead, New York, USA; and Microarray Core Facility, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
37
|
Rosenfeld SM, Perry HM, Gonen A, Prohaska TA, Srikakulapu P, Grewal S, Das D, McSkimming C, Taylor AM, Tsimikas S, Bender TP, Witztum JL, McNamara CA. B-1b Cells Secrete Atheroprotective IgM and Attenuate Atherosclerosis. Circ Res 2015; 117:e28-39. [PMID: 26082558 DOI: 10.1161/circresaha.117.306044] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 06/16/2015] [Indexed: 01/28/2023]
Abstract
RATIONALE B cells contribute to atherosclerosis through subset-specific mechanisms. Whereas some controversy exists about the role of B-2 cells, B-1a cells are atheroprotective because of secretion of atheroprotective IgM antibodies independent of antigen. B-1b cells, a unique subset of B-1 cells that respond specifically to T-cell-independent antigens, have not been studied within the context of atherosclerosis. OBJECTIVE To determine whether B-1b cells produce atheroprotective IgM antibodies and function to protect against diet-induced atherosclerosis. METHODS AND RESULTS We demonstrate that B-1b cells are sufficient to produce IgM antibodies against oxidation-specific epitopes on low-density lipoprotein both in vitro and in vivo. In addition, we demonstrate that B-1b cells provide atheroprotection after adoptive transfer into B- and T-cell deficient (Rag1(-/-)Apoe(-/-)) hosts. We implicate inhibitor of differentiation 3 (Id3) in the regulation of B-1b cells as B-cell-specific Id3 knockout mice (Id3(BKO)Apoe(-/-)) have increased numbers of B-1b cells systemically, increased titers of oxidation-specific epitope-reactive IgM antibodies, and significantly reduced diet-induced atherosclerosis when compared with Id3(WT)Apoe(-/-) controls. Finally, we report that the presence of a homozygous single nucleotide polymorphism in ID3 in humans that attenuates Id3 function is associated with an increased percentage of circulating B-1 cells and anti-malondialdehyde-low-density lipoprotein IgM suggesting clinical relevance. CONCLUSIONS These results provide novel evidence that B-1b cells produce atheroprotective oxidation-specific epitope-reactive IgM antibodies and protect against atherosclerosis in mice and suggest that similar mechanisms may occur in humans.
Collapse
Affiliation(s)
- Sam M Rosenfeld
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Heather M Perry
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Ayelet Gonen
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Thomas A Prohaska
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Prasad Srikakulapu
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Sukhdeep Grewal
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Deepanjana Das
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Chantel McSkimming
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Angela M Taylor
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Sotirios Tsimikas
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Timothy P Bender
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Joseph L Witztum
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla
| | - Coleen A McNamara
- From the Cardiovascular Research Center (S.M.R., H.M.P., P.S., S.G., D.D., C.M., C.A.M.), Department of Pathology (S.M.R., H.M.P.), Department of Medicine, Division of Cardiovascular Medicine (A.M.T., C.A.M.), and Beirne B. Carter Center for Immunology Research (T.P.B., C.A.M.), University of Virginia, Charlottesville; and Department of Medicine, Division of Endocrinology and Metabolism (A.G., T.A.P., J.L.W.) and Department of Medicine, Division of Cardiology (S.T.), University of California San Diego, La Jolla.
| |
Collapse
|
38
|
Gao H, Bu Y, Wu Q, Wang X, Chang N, Lei L, Chen S, Liu D, Zhu X, Hu K, Xiong JW. Mecp2 regulates neural cell differentiation by suppressing the Id1 to Her2 axis in zebrafish. J Cell Sci 2015; 128:2340-50. [DOI: 10.1242/jcs.167874] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/28/2015] [Indexed: 01/20/2023] Open
Abstract
ABSTRACT
Rett syndrome (RTT) is a progressive neurological disorder caused by mutations in the X-linked protein methyl-CpG-binding protein 2 (MeCP2). The endogenous function of MeCP2 during neural differentiation is still unclear. Here, we report that mecp2 is required for brain development in zebrafish. Mecp2 was broadly expressed initially in embryos and enriched later in the brain. Either morpholino knockdown or genetic depletion of mecp2 inhibited neuronal differentiation, whereas its overexpression promoted neuronal differentiation, suggesting an essential role of mecp2 in directing neural precursors into differentiated neurons. Mechanistically, her2 (the zebrafish ortholog of mammalian Hes5) was upregulated in mecp2 morphants in an Id1-dependent manner. Moreover, knockdown of either her2 or id1 fully rescued neuronal differentiation in mecp2 morphants. These results suggest that Mecp2 plays an important role in neural cell development by suppressing the Id1–Her2 axis, and provide new evidence that embryonic neural defects contribute to the later motor and cognitive dysfunctions in RTT.
Collapse
Affiliation(s)
- Hai Gao
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Ye Bu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Qing Wu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xu Wang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Nannan Chang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Lei Lei
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Shilin Chen
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Dong Liu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Life Sciences, Peking University, Beijing, China
| | - Xiaojun Zhu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Keping Hu
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Jing-Wei Xiong
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, China
| |
Collapse
|
39
|
Mele F, Basso C, Leoni C, Aschenbrenner D, Becattini S, Latorre D, Lanzavecchia A, Sallusto F, Monticelli S. ERK phosphorylation and miR-181a expression modulate activation of human memory TH17 cells. Nat Commun 2015; 6:6431. [DOI: 10.1038/ncomms7431] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 01/28/2015] [Indexed: 12/19/2022] Open
|
40
|
Abstract
Adaptive immunity is involved in the pathogenesis of atherosclerosis, but the recruitment of T and B lymphocytes to atherosclerotic lesions is not as well studied as that of monocytes. In this review, we summarize the current understanding of the role of lymphocyte subsets in the pathogenesis of atherosclerosis and discuss chemokines and chemokine receptors involved in lymphocyte homing to atherosclerotic lesions. We review evidence for involvement of the chemokines CCL5, CCL19, CCL21, CXCL10, and CXCL16 and macrophage migration inhibitory factor in lymphocyte homing in atherosclerosis. Also, we review the role of their receptors CCR5, CCR6, CCR7, CXCR3, CXCR6, and CXCR2/CXCR4 and the role of the L-selectin in mouse models of atherosclerosis.
Collapse
Affiliation(s)
- Jie Li
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA
| | - Klaus Ley
- From the Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, CA.
| |
Collapse
|
41
|
Belle I, Mahlios J, McKenzie A, Zhuang Y. Aberrant production of IL-13 by T cells promotes exocrinopathy in Id3 knockout mice. Cytokine 2014; 69:226-33. [PMID: 25010390 DOI: 10.1016/j.cyto.2014.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 04/24/2014] [Accepted: 06/11/2014] [Indexed: 12/24/2022]
Abstract
Elevated levels of the cytokine IL-13 has been found to be associated with autoimmune diseases, including Sjögren's Syndrome. However, whether IL-13 plays a causative role in disease development is not known and cannot be easily studied in humans. Our previous work has shown that levels of IL-13 are elevated in Id3 knockout mice, which has been established as a model for primary Sjögren's Syndrome. Here, we utilized an IL-13 reporter to determine the source of the elevated IL-13 levels observed in Id3 knockout mice and assess its contribution to SS pathology. Our results indicate that T cells, notably CD4 and γδ T cells, in Id3 knockout mice acquire IL-13 competency at an elevated rate well before disease symptoms become apparent. We also show that T cells developing early in life are more predisposed to produce IL-13. Finally, analysis of Id3 and IL-13 double deficient mice demonstrated that IL-13 plays an essential role in the deterioration of gland function. Our study provides crucial genetic evidence that enhanced IL-13 production by T cells can play a causative role in the exocrinopathy observed in Id3 knockout mice.
Collapse
Affiliation(s)
- Ian Belle
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Josh Mahlios
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Yuan Zhuang
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
42
|
Expression and recruitment of uracil-DNA glycosylase are regulated by E2A during antibody diversification. Mol Immunol 2014; 60:23-31. [DOI: 10.1016/j.molimm.2014.03.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 03/24/2014] [Indexed: 11/20/2022]
|
43
|
Lasorella A, Benezra R, Iavarone A. The ID proteins: master regulators of cancer stem cells and tumour aggressiveness. Nat Rev Cancer 2014; 14:77-91. [PMID: 24442143 DOI: 10.1038/nrc3638] [Citation(s) in RCA: 267] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inhibitor of DNA binding (ID) proteins are transcriptional regulators that control the timing of cell fate determination and differentiation in stem and progenitor cells during normal development and adult life. ID genes are frequently deregulated in many types of human neoplasms, and they endow cancer cells with biological features that are hijacked from normal stem cells. The ability of ID proteins to function as central 'hubs' for the coordination of multiple cancer hallmarks has established these transcriptional regulators as therapeutic targets and biomarkers in specific types of human tumours.
Collapse
Affiliation(s)
- Anna Lasorella
- Institute for Cancer Genetics, Department of Pathology and Pediatrics, Columbia University Medical Center, 1130 St. Nicholas Avenue, New York, 10032 New York, USA
| | - Robert Benezra
- Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 241, New York, 10065 New York, USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Department of Pathology and Neurology, Columbia University Medical Center, 1130 St. Nicholas Avenue, New York, 10032 New York, USA
| |
Collapse
|
44
|
Abstract
As members of the basic helix-loop-helix (bHLH) family of transcription factors, E proteins function in the immune system by directing and maintaining a vast transcriptional network that regulates cell survival, proliferation, differentiation, and function. Proper activity of this network is essential to the functionality of the immune system. Aberrations in E protein expression or function can cause numerous defects, ranging from impaired lymphocyte development and immunodeficiency to aberrant function, cancer, and autoimmunity. Additionally, disruption of inhibitor of DNA-binding (Id) proteins, natural inhibitors of E proteins, can induce additional defects in development and function. Although E proteins have been investigated for several decades, their study continues to yield novel and exciting insights into the workings of the immune system. The goal of this chapter is to discuss the various classical roles of E proteins in lymphocyte development and highlight new and ongoing research into how these roles, if compromised, can lead to disease.
Collapse
Affiliation(s)
- Ian Belle
- Department of Immunology, Duke University Medical Center, Durham North Carolina, USA.
| | - Yuan Zhuang
- Department of Immunology, Duke University Medical Center, Durham North Carolina, USA
| |
Collapse
|
45
|
Zhang B, Lin YY, Dai M, Zhuang Y. Id3 and Id2 act as a dual safety mechanism in regulating the development and population size of innate-like γδ T cells. THE JOURNAL OF IMMUNOLOGY 2013; 192:1055-1063. [PMID: 24379125 DOI: 10.4049/jimmunol.1302694] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The innate-like T cells expressing Vγ1.1 and Vδ6.3 represent a unique T cell lineage sharing features with both the γδ T and the invariant NKT cells. The population size of Vγ1.1(+)Vδ6.3(+) T cells is tightly controlled and usually contributes to a very small proportion of thymic output, but the underlying mechanism remains enigmatic. Deletion of Id3, an inhibitor of E protein transcription factors, can induce an expansion of the Vγ1.1(+)Vδ6.3(+) T cell population. This phenotype is much stronger on the C57BL/6 background than on the 129/sv background. Using quantitative trait linkage analysis, we identified Id2, a homolog of Id3, to be the major modifier of Id3 in limiting Vγ1.1(+)Vδ6.3(+) T cell expansion. The Vγ1.1(+)Vδ6.3(+) phenotype is attributed to an intrinsic weakness of Id2 transcription from Id2 C57BL/6 allele, leading to an overall reduced dosage of Id proteins. However, complete removal of both Id2 and Id3 genes in developing T cells suppressed the expansion of Vγ1.1(+)Vδ6.3(+) T cells because of decreased proliferation and increased cell death. We showed that conditional knockout of Id2 alone is sufficient to promote a moderate expansion of γδ T cells. These regulatory effects of Id2 and Id3 on Vγ1.1(+)Vδ6.3(+) T cells are mediated by titration of E protein activity, because removing one or more copies of E protein genes can restore Vγ1.1(+)Vδ6.3(+) T cell expansion in Id2 and Id3 double conditional knockout mice. Our data indicated that Id2 and Id3 collaboratively control survival and expansion of the γδ lineage through modulating a proper threshold of E proteins.
Collapse
Affiliation(s)
- Baojun Zhang
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yen-Yu Lin
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Meifang Dai
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yuan Zhuang
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
46
|
Verykokakis M, Krishnamoorthy V, Iavarone A, Lasorella A, Sigvardsson M, Kee BL. Essential functions for ID proteins at multiple checkpoints in invariant NKT cell development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:5973-83. [PMID: 24244015 PMCID: PMC3864619 DOI: 10.4049/jimmunol.1301521] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Invariant NKT (iNKT) cells display characteristics of both adaptive and innate lymphoid cells (ILCs). Like other ILCs, iNKT cells constitutively express ID proteins, which antagonize the E protein transcription factors that are essential for adaptive lymphocyte development. However, unlike ILCs, ID2 is not essential for thymic iNKT cell development. In this study, we demonstrated that ID2 and ID3 redundantly promoted iNKT cell lineage specification involving the induction of the signature transcription factor PLZF and that ID3 was critical for development of TBET-dependent NKT1 cells. In contrast, both ID2 and ID3 limited iNKT cell numbers by enforcing the postselection checkpoint in conventional thymocytes. Therefore, iNKT cells show both adaptive and innate-like requirements for ID proteins at distinct checkpoints during iNKT cell development.
Collapse
Affiliation(s)
- Mihalis Verykokakis
- Department of Pathology, University of Chicago, Chicago, IL, 60637
- Committee on Immunology, University of Chicago, Chicago, IL, 60637
| | | | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, 10032
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032
- Department of Pathology, Columbia University Medical Center, New York, NY, 10032
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, 10032
- Department of Neurology, Columbia University Medical Center, New York, NY, 10032
- Department of Pathology, Columbia University Medical Center, New York, NY, 10032
| | - Mikael Sigvardsson
- Department of Clinical and Experimental Medicine, Experimental Hematopoiesis Unit, Faculty for Health Sciences, Linköping University, 58183 Linköping, Sweden
| | - Barbara L. Kee
- Department of Pathology, University of Chicago, Chicago, IL, 60637
- Committee on Immunology, University of Chicago, Chicago, IL, 60637
| |
Collapse
|
47
|
Perry HM, Oldham SN, Fahl SP, Que X, Gonen A, Harmon DB, Tsimikas S, Witztum JL, Bender TP, McNamara CA. Helix-loop-helix factor inhibitor of differentiation 3 regulates interleukin-5 expression and B-1a B cell proliferation. Arterioscler Thromb Vasc Biol 2013; 33:2771-9. [PMID: 24115031 DOI: 10.1161/atvbaha.113.302571] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Natural immunity is emerging as an important mediator of protection from atherogenesis. Natural IgM antibodies that recognize oxidation-specific epitopes on low-density lipoprotein or phospholipids and the B-1a B cells that produce them attenuate atherosclerosis. We previously demonstrated that Apoe(-/-) mice globally deficient in the helix-loop-helix protein inhibitor of differentiation 3 (Id3) develop early diet-induced atherosclerosis. Furthermore, B cell-mediated attenuation of atherosclerosis in B cell-deficient mice was dependent on Id3. Here, we sought to determine whether Id3 regulates B-1a B cells and the natural antibodies that they produce and identify mechanisms mediating these effects. APPROACH AND RESULTS Mice lacking Id3 had significantly fewer B-1a B cells in the spleen and peritoneal cavity and reduced serum levels of the natural antibody E06. B cell-specific deletion of Id3 revealed that this effect was not because of the loss of Id3 in B cells. Interleukin (IL)-33 induced abundant, Id3-dependent IL-5 production in the recently identified innate lymphoid cell, the natural helper (NH) cell, but not Th2 or mast cells. In addition, delivery of IL-5 to Id3-deficient mice restored B-1a B cell proliferation. B-1a B cells were present in aortic samples also containing NH cells. Aortic NH cells produced IL-5, a B-1a B cell mitogen in response to IL-33 stimulation. CONCLUSIONS These studies are the first to identify NH and B-1a B cells in the aorta and provide evidence that Id3 is a key regulator of NH cell IL-5 production and B-1a B cell homeostasis.
Collapse
Affiliation(s)
- Heather M Perry
- From the Cardiovascular Research Center (H.M.P., S.N.O., D.B.H., C.A.M.), Department of Pathology (H.M.P.), Department of Medicine (S.N.O.), Beirne B. Carter Center for Immunology Research (S.P.F., C.A.M.), Department of Microbiology, Immunology and Cancer Biology (S.P.F., T.P.B.), Department of Biochemistry, Molecular Biology and Genetics (D.B.H., T.P.B.), Department of Medicine, Cardiovascular Division (C.A.M.), Department of Molecular Physiology and Biological Physics (C.A.M.), University of Virginia, Charlottesville; and the Department of Medicine, University of California, San Diego (X.Q., A.G., S.T., J.L.W.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Sharma P, Knowell AE, Chinaranagari S, Komaragiri S, Nagappan P, Patel D, Havrda MC, Chaudhary J. Id4 deficiency attenuates prostate development and promotes PIN-like lesions by regulating androgen receptor activity and expression of NKX3.1 and PTEN. Mol Cancer 2013; 12:67. [PMID: 23786676 PMCID: PMC3694449 DOI: 10.1186/1476-4598-12-67] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 06/12/2013] [Indexed: 12/30/2022] Open
Abstract
Background Inhibitor of differentiation 4 (Id4), a member of the helix-loop-helix family of transcriptional regulators has emerged as a tumor suppressor in prostate cancer. Id4 is expressed in the normal prostate where its expression is also regulated by androgens. In this study we investigated the effect of loss of Id4 (Id4-/-) on adult prostate morphology. Methods Histological analysis was performed on prostates from 6-8 weeks old Id4-/-, Id4+/- and Id4+/+ mice. Expression of Id1, Sox9, Myc, androgen receptor, Akt, p-Akt, Pten and Nkx3.1 was investigated by immunohistochemistry. Androgen receptor binding on NKX3.1 promoter was studied by chromatin immuno-precipitation. Id4 was either over-expressed or silenced in prostate cancer cell lines DU145 and LNCaP respectively followed by analysis of PTEN, NKX3.1 and Sox9 expression. Results Id4-/- mice had smaller prostates with fewer tubules, smaller tubule diameters and subtle mPIN like lesions. Levels of androgen receptor were similar between wild type and Id4-/- prostate. Decreased NKX3.1 expression was in part due to decreased androgen receptor binding on NKX3.1 promoter in Id4-/- mice. The increase in the expression of Myc, Sox9, Id1, Ki67 and decrease in the expression of PTEN, Akt and phospho-AKT was associated with subtle mPIN like lesions in Id4-/- prostates. Finally, prostate cancer cell line models in which Id4 was either silenced or over-expressed confirmed that Id4 regulates NKX3.1, Sox9 and PTEN. Conclusions Our results suggest that loss of Id4 attenuates normal prostate development and promotes hyperplasia/dysplasia with subtle mPIN like lesions characterized by gain of Myc and Id1 and loss of Nkx3.1 and Pten expression. One of the mechanisms by which Id4 may regulate normal prostate development is through regulating androgen receptor binding to respective response elements such as those on NKX3.1 promoter. In spite of these complex alterations, large neoplastic lesions in Id4-/- prostates were not observed suggesting the possibility of mechanisms/pathways such as loss of Akt that could restrain the formation of significant pre-cancerous lesions.
Collapse
|
49
|
Furman D, Jojic V, Kidd B, Shen-Orr S, Price J, Jarrell J, Tse T, Huang H, Lund P, Maecker HT, Utz PJ, Dekker CL, Koller D, Davis MM. Apoptosis and other immune biomarkers predict influenza vaccine responsiveness. Mol Syst Biol 2013; 9:659. [PMID: 23591775 PMCID: PMC3658270 DOI: 10.1038/msb.2013.15] [Citation(s) in RCA: 145] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 03/07/2013] [Indexed: 12/17/2022] Open
Abstract
Despite the importance of the immune system in many diseases, there are currently no objective benchmarks of immunological health. In an effort to identifying such markers, we used influenza vaccination in 30 young (20-30 years) and 59 older subjects (60 to >89 years) as models for strong and weak immune responses, respectively, and assayed their serological responses to influenza strains as well as a wide variety of other parameters, including gene expression, antibodies to hemagglutinin peptides, serum cytokines, cell subset phenotypes and in vitro cytokine stimulation. Using machine learning, we identified nine variables that predict the antibody response with 84% accuracy. Two of these variables are involved in apoptosis, which positively associated with the response to vaccination and was confirmed to be a contributor to vaccine responsiveness in mice. The identification of these biomarkers provides new insights into what immune features may be most important for immune health.
Collapse
Affiliation(s)
- David Furman
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Vladimir Jojic
- Department of Computer Science, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Brian Kidd
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Shai Shen-Orr
- Department of Immunology, Faculty of Medicine, Technion, Technion City, Haifa, Israel
| | - Jordan Price
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Justin Jarrell
- Division of Immunology and Rheumatology, Department of Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Tiffany Tse
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Huang Huang
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Peder Lund
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Paul J Utz
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
- Division of Immunology and Rheumatology, Department of Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Cornelia L Dekker
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
- Department of Pediatrics, Division of Infectious Diseases, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Daphne Koller
- Department of Computer Science, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, School of Medicine, Stanford University, Palo Alto, CA, USA
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Palo Alto, CA, USA
- The Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
50
|
Niola F, Zhao X, Singh D, Sullivan R, Castano A, Verrico A, Zoppoli P, Friedmann-Morvinski D, Sulman E, Barrett L, Zhuang Y, Verma I, Benezra R, Aldape K, Iavarone A, Lasorella A. Mesenchymal high-grade glioma is maintained by the ID-RAP1 axis. J Clin Invest 2012; 123:405-17. [PMID: 23241957 DOI: 10.1172/jci63811] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 10/11/2012] [Indexed: 01/10/2023] Open
Abstract
High-grade gliomas (HGGs) are incurable brain tumors that are characterized by the presence of glioma-initiating cells (GICs). GICs are essential to tumor aggressiveness and retain the capacity for self-renewal and multilineage differentiation as long as they reside in the perivascular niche. ID proteins are master regulators of stemness and anchorage to the extracellular niche microenvironment, suggesting that they may play a role in maintaining GICs. Here, we modeled the probable therapeutic impact of ID inactivation in HGG by selective ablation of Id in tumor cells and after tumor initiation in a new mouse model of human mesenchymal HGG. Deletion of 3 Id genes induced rapid release of GICs from the perivascular niche, followed by tumor regression. GIC displacement was mediated by derepression of Rap1gap and subsequent inhibition of RAP1, a master regulator of cell adhesion. We identified a signature module of 5 genes in the ID pathway, including RAP1GAP, which segregated 2 subgroups of glioma patients with markedly different clinical outcomes. The model-informed survival analysis together with genetic and functional studies establish that ID activity is required for the maintenance of mesenchymal HGG and suggest that pharmacological inactivation of ID proteins could serve as a therapeutic strategy.
Collapse
Affiliation(s)
- Francesco Niola
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|