1
|
Weiss ES, Hirai T, Li H, Liu A, Baker S, Magill I, Gillis J, Zhang YR, Ramcke T, Kurihara K, Masopust D, Anandasabapathy N, Singh H, Zemmour D, Mackay LK, Kaplan DH. Epidermal Resident Memory T Cell Fitness Requires Antigen Encounter in the Skin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646438. [PMID: 40236062 PMCID: PMC11996394 DOI: 10.1101/2025.03.31.646438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
CD8 + tissue resident memory T cells (T RM ) develop from effectors that seed peripheral tissues where they persist providing defense against subsequent challenges. T RM persistence requires autocrine TGFβ transactivated by integrins expressed on keratinocytes. T RM precursors that encounter antigen in the epidermis during development outcompete bystander T RM for TGFβ resulting in enhanced persistence. ScRNA-seq analysis of epidermal T RM revealed that local antigen experience in the skin resulted in an enhanced differentiation signature in comparison with bystanders. Upon recall, T RM displayed greater proliferation dictated by affinity of antigen experienced during epidermal development. Finally, local antigen experienced T RM differentially expressed TGFβRIII, which increases avidity of the TGFβRI/II receptor complex for TGFβ. Selective ablation of Tgfbr3 reduced local antigen experienced T RM capacity to persist, rendering them phenotypically like bystander T RM . Thus, antigen driven TCR signaling in the epidermis during T RM differentiation results in a lower TGFβ requirement for persistence and increased proliferative capacity that together enhance epidermal T RM fitness.
Collapse
|
2
|
Osum KC, Becker SH, Krueger PD, Mitchell JS, Hong SW, Magill IR, Jenkins MK. A minority of Th1 and Tfh effector cells express survival genes shared by memory cell progeny that require IL-7 or TCR signaling to persist. Cell Rep 2025; 44:115111. [PMID: 39723889 PMCID: PMC12009130 DOI: 10.1016/j.celrep.2024.115111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 10/24/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
It is not clear how CD4+ memory T cells are formed from a much larger pool of earlier effector cells. We found that transient systemic bacterial infection rapidly generates several antigen-specific T helper (Th)1 and T follicular helper (Tfh) cell populations with different tissue residence behaviors. Although most cells of all varieties had transcriptomes indicative of cell stress and death at the peak of the response, some had already acquired a memory cell signature characterized by expression of genes involved in cell survival. Each Th1 and Tfh cell type was maintained long term by interleukin (IL)-7, except germinal center Tfh cells, which depended on a T cell antigen receptor (TCR) signal. The results indicate that acute infection induces rapid differentiation of Th1 and Tfh cells, a minority of which quickly adopt the gene expression profile of memory cells and survive by signals from the IL-7 receptor or TCR.
Collapse
Affiliation(s)
- Kevin C Osum
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Samuel H Becker
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Peter D Krueger
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Jason S Mitchell
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Sung-Wook Hong
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Biotechnology, Yonsei University, Seoul, South Korea
| | - Ian R Magill
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Marc K Jenkins
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
3
|
Chen W, Kim S, Kim SY, Beheshtian C, Kim N, Shin KH, Kim RH, Kim S, Park NH. GV1001, hTERT Peptide Fragment, Prevents Doxorubicin-Induced Endothelial-to-Mesenchymal Transition in Human Endothelial Cells and Atherosclerosis in Mice. Cells 2025; 14:98. [PMID: 39851526 PMCID: PMC11763685 DOI: 10.3390/cells14020098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Doxorubicin is a highly effective anticancer agent, but its clinical use is restricted by severe side effects, including atherosclerosis and cardiomyopathy. These complications are partly attributed to doxorubicin's ability to induce endothelial-to-mesenchymal transition (EndMT) in vascular endothelial cells, a critical process in the initiation and progression of atherosclerosis and cardiomyopathy. GV1001, a multifunctional peptide with anti-inflammatory, anti-cancer, antioxidant, and anti-Alzheimer's properties, has demonstrated inhibition of EndMT. We investigated whether GV1001 could counteract doxorubicin-induced EndMT in endothelial cells and prevent atherosclerosis in a mouse model. The results revealed that GV1001 significantly suppressed EndMT induced by doxorubicin, likely through its protective effects on mitochondria. By mitigating mitochondrial damage, GV1001 reduced the accumulation of mitochondrial and cellular reactive oxygen species (ROS), repressed the activation of nuclear factor kappa B (NF-κB), and reduced the production of proinflammatory cytokines in endothelial cells. Additionally, GV1001 reduced systemic and vascular inflammation, lipid accumulation, and monocyte/macrophage infiltration within arterial walls in mice. In conclusion, GV1001 appears to prevent doxorubicin-induced atherosclerosis by safeguarding vascular endothelial cells from mitochondrial dysfunction, inflammation, and phenotypic changes. These findings suggest the potential of GV1001 as a therapeutic agent to mitigate the long-term cardiovascular side effects associated with doxorubicin treatment in humans.
Collapse
Affiliation(s)
- Wei Chen
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, University of California, 714 Tiverton Ave, Los Angeles, CA 90095, USA; (W.C.); (S.K.); (S.Y.K.); (C.B.); (N.K.); (K.-H.S.); (R.H.K.)
| | - Seojin Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, University of California, 714 Tiverton Ave, Los Angeles, CA 90095, USA; (W.C.); (S.K.); (S.Y.K.); (C.B.); (N.K.); (K.-H.S.); (R.H.K.)
| | - Sharon Y. Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, University of California, 714 Tiverton Ave, Los Angeles, CA 90095, USA; (W.C.); (S.K.); (S.Y.K.); (C.B.); (N.K.); (K.-H.S.); (R.H.K.)
| | - Cheyenne Beheshtian
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, University of California, 714 Tiverton Ave, Los Angeles, CA 90095, USA; (W.C.); (S.K.); (S.Y.K.); (C.B.); (N.K.); (K.-H.S.); (R.H.K.)
| | - Naryung Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, University of California, 714 Tiverton Ave, Los Angeles, CA 90095, USA; (W.C.); (S.K.); (S.Y.K.); (C.B.); (N.K.); (K.-H.S.); (R.H.K.)
| | - Ki-Hyuk Shin
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, University of California, 714 Tiverton Ave, Los Angeles, CA 90095, USA; (W.C.); (S.K.); (S.Y.K.); (C.B.); (N.K.); (K.-H.S.); (R.H.K.)
- UCLA Jonsson Comprehensive Cancer Center, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - Reuben H. Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, University of California, 714 Tiverton Ave, Los Angeles, CA 90095, USA; (W.C.); (S.K.); (S.Y.K.); (C.B.); (N.K.); (K.-H.S.); (R.H.K.)
- UCLA Jonsson Comprehensive Cancer Center, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | - Sangjae Kim
- Teloid Inc., 920 Westholme Avenue, Los Angeles, CA 90024, USA
| | - No-Hee Park
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, University of California, 714 Tiverton Ave, Los Angeles, CA 90095, USA; (W.C.); (S.K.); (S.Y.K.); (C.B.); (N.K.); (K.-H.S.); (R.H.K.)
- UCLA Jonsson Comprehensive Cancer Center, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, University of California, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
Li W, Huang Y, Liu J, Zhou Y, Sun H, Fan Y, Liu F. Defective macrophage efferocytosis in advanced atherosclerotic plaque and mitochondrial therapy. Life Sci 2024; 359:123204. [PMID: 39491771 DOI: 10.1016/j.lfs.2024.123204] [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: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease primarily affecting large and medium-sized arterial vessels, characterized by lipoprotein disorders, intimal thickening, smooth muscle cell proliferation, and the formation of vulnerable plaques. Macrophages (MΦs) play a vital role in the inflammatory response throughout all stages of atherosclerotic development and are considered significant therapeutic targets. In early lesions, macrophage efferocytosis rapidly eliminates harmful cells. However, impaired efferocytosis in advanced plaques perpetuates the inflammatory microenvironment of AS. Defective efferocytosis has emerged as a key factor in atherosclerotic pathogenesis and the progression to severe cardiovascular disease. Herein, this review probes into investigate the potential mechanisms at the cellular, molecular, and organelle levels underlying defective macrophage efferocytosis in advanced lesion plaques. In the inflammatory microenvironments of AS with interactions among diverse inflammatory immune cells, impaired macrophage efferocytosis is strongly linked to multiple factors, such as a lower absolute number of phagocytes, the aberrant expression of crucial molecules, and impaired mitochondrial energy provision in phagocytes. Thus, focusing on molecular targets to enhance macrophage efferocytosis or targeting mitochondrial therapy to restore macrophage metabolism homeostasis has emerged as a potential strategy to mitigate the progression of advanced atherosclerotic plaque, providing various treatment options.
Collapse
Affiliation(s)
- Wanling Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yaqing Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Hongyu Sun
- The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yonghong Fan
- The General Hospital of Western Theater Command, Chengdu 610083, China.
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| |
Collapse
|
5
|
Sankaran DG, Zhu H, Maymi VI, Forlastro IM, Jiang Y, Laniewski N, Scheible KM, Rudd BD, Grimson AW. Gene Regulatory Programs that Specify Age-Related Differences during Thymocyte Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.599011. [PMID: 38948840 PMCID: PMC11212896 DOI: 10.1101/2024.06.14.599011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
T cell development is fundamental to immune system establishment, yet how this development changes with age remains poorly understood. Here, we construct a transcriptional and epigenetic atlas of T cell developmental programs in neonatal and adult mice, revealing the ontogeny of divergent gene regulatory programs and their link to age-related differences in phenotype and function. Specifically, we identify a gene module that diverges with age from the earliest stages of genesis and includes programs that govern effector response and cell cycle regulation. Moreover, we reveal that neonates possess more accessible chromatin during early thymocyte development, likely establishing poised gene expression programs that manifest later in thymocyte development. Finally, we leverage this atlas, employing a CRISPR-based perturbation approach coupled with single-cell RNA sequencing as a readout to uncover a conserved transcriptional regulator, Zbtb20, that contributes to age-dependent differences in T cell development. Altogether, our study defines transcriptional and epigenetic programs that regulate age-specific differences in T cell development.
Collapse
|
6
|
Gatti DM, Tyler AL, Mahoney JM, Churchill GA, Yener B, Koyuncu D, Gurcan MN, Niazi MKK, Tavolara T, Gower A, Dayao D, McGlone E, Ginese ML, Specht A, Alsharaydeh A, Tessier PA, Kurtz SL, Elkins KL, Kramnik I, Beamer G. Systems genetics uncover new loci containing functional gene candidates in Mycobacterium tuberculosis-infected Diversity Outbred mice. PLoS Pathog 2024; 20:e1011915. [PMID: 38861581 PMCID: PMC11195971 DOI: 10.1371/journal.ppat.1011915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/24/2024] [Accepted: 04/17/2024] [Indexed: 06/13/2024] Open
Abstract
Mycobacterium tuberculosis infects two billion people across the globe, and results in 8-9 million new tuberculosis (TB) cases and 1-1.5 million deaths each year. Most patients have no known genetic basis that predisposes them to disease. Here, we investigate the complex genetic basis of pulmonary TB by modelling human genetic diversity with the Diversity Outbred mouse population. When infected with M. tuberculosis, one-third develop early onset, rapidly progressive, necrotizing granulomas and succumb within 60 days. The remaining develop non-necrotizing granulomas and survive longer than 60 days. Genetic mapping using immune and inflammatory mediators; and clinical, microbiological, and granuloma correlates of disease identified five new loci on mouse chromosomes 1, 2, 4, 16; and three known loci on chromosomes 3 and 17. Further, multiple positively correlated traits shared loci on chromosomes 1, 16, and 17 and had similar patterns of allele effects, suggesting these loci contain critical genetic regulators of inflammatory responses to M. tuberculosis. To narrow the list of candidate genes, we used a machine learning strategy that integrated gene expression signatures from lungs of M. tuberculosis-infected Diversity Outbred mice with gene interaction networks to generate scores representing functional relationships. The scores were used to rank candidates for each mapped trait, resulting in 11 candidate genes: Ncf2, Fam20b, S100a8, S100a9, Itgb5, Fstl1, Zbtb20, Ddr1, Ier3, Vegfa, and Zfp318. Although all candidates have roles in infection, inflammation, cell migration, extracellular matrix remodeling, or intracellular signaling, and all contain single nucleotide polymorphisms (SNPs), SNPs in only four genes (S100a8, Itgb5, Fstl1, Zfp318) are predicted to have deleterious effects on protein functions. We performed methodological and candidate validations to (i) assess biological relevance of predicted allele effects by showing that Diversity Outbred mice carrying PWK/PhJ alleles at the H-2 locus on chromosome 17 QTL have shorter survival; (ii) confirm accuracy of predicted allele effects by quantifying S100A8 protein in inbred founder strains; and (iii) infection of C57BL/6 mice deficient for the S100a8 gene. Overall, this body of work demonstrates that systems genetics using Diversity Outbred mice can identify new (and known) QTLs and functionally relevant gene candidates that may be major regulators of complex host-pathogens interactions contributing to granuloma necrosis and acute inflammation in pulmonary TB.
Collapse
Affiliation(s)
- Daniel M. Gatti
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Anna L. Tyler
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | | | | | - Bulent Yener
- Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Deniz Koyuncu
- Rensselaer Polytechnic Institute, Troy, New York, United States of America
| | - Metin N. Gurcan
- Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - MK Khalid Niazi
- Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - Thomas Tavolara
- Wake Forest University School of Medicine, Winston Salem, North Carolina, United States of America
| | - Adam Gower
- Clinical and Translational Science Institute, Boston University, Boston, Massachusetts, United States of America
| | - Denise Dayao
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Emily McGlone
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Melanie L. Ginese
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Aubrey Specht
- Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Anas Alsharaydeh
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Philipe A. Tessier
- Department of Microbiology and Immunology, Laval University School of Medicine, Quebec, Canada
| | - Sherry L. Kurtz
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Karen L. Elkins
- Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Igor Kramnik
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, United States of America
| | - Gillian Beamer
- Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| |
Collapse
|
7
|
Tura A, Herfs V, Maaßen T, Zuo H, Vardanyan S, Prasuhn M, Ranjbar M, Kakkassery V, Grisanti S. Quercetin Impairs the Growth of Uveal Melanoma Cells by Interfering with Glucose Uptake and Metabolism. Int J Mol Sci 2024; 25:4292. [PMID: 38673877 PMCID: PMC11049862 DOI: 10.3390/ijms25084292] [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: 03/13/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Monosomy 3 in uveal melanoma (UM) increases the risk of lethal metastases, mainly in the liver, which serves as the major site for the storage of excessive glucose and the metabolization of the dietary flavonoid quercetin. Although primary UMs with monosomy 3 exhibit a higher potential for basal glucose uptake, it remains unknown as to whether glycolytic capacity is altered in such tumors. Herein, we initially analyzed the expression of n = 151 genes involved in glycolysis and its interconnected branch, the "pentose phosphate pathway (PPP)", in the UM cohort of The Cancer Genome Atlas Study and validated the differentially expressed genes in two independent cohorts. We also evaluated the effects of quercetin on the growth, survival, and glucose metabolism of the UM cell line 92.1. The rate-limiting glycolytic enzyme PFKP was overexpressed whereas the ZBTB20 gene (locus: 3q13.31) was downregulated in the patients with metastases in all cohorts. Quercetin was able to impair proliferation, viability, glucose uptake, glycolysis, ATP synthesis, and PPP rate-limiting enzyme activity while increasing oxidative stress. UMs with monosomy 3 display a stronger potential to utilize glucose for the generation of energy and biomass. Quercetin can prevent the growth of UM cells by interfering with glucose metabolism.
Collapse
Affiliation(s)
- Aysegül Tura
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23562 Luebeck, Germany; (V.H.); (T.M.); (H.Z.); (S.V.); (M.P.); (V.K.); (S.G.)
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Liu J, Zhang H. Zinc Finger and BTB Domain-Containing 20: A Newly Emerging Player in Pathogenesis and Development of Human Cancers. Biomolecules 2024; 14:192. [PMID: 38397429 PMCID: PMC10887282 DOI: 10.3390/biom14020192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Zinc finger and BTB domain-containing 20 (ZBTB20), which was initially identified in human dendritic cells, belongs to a family of transcription factors (TFs) with an N-terminal BTB domain and one or more C-terminal DNA-binding zinc finger domains. Under physiological conditions, ZBTB20 acts as a transcriptional repressor in cellular development and differentiation, metabolism, and innate immunity. Interestingly, multiple lines of evidence from mice and human systems have revealed the importance of ZBTB20 in the pathogenesis and development of cancers. ZBTB20 is not only a hotspot of genetic variation or fusion in many types of human cancers, but also a key TF or intermediator involving in the dysregulation of cancer cells. Given the diverse functions of ZBTB20 in both health and disease, we herein summarize the structure and physiological roles of ZBTB20, with an emphasis on the latest findings on tumorigenesis and cancer progression.
Collapse
Affiliation(s)
| | - Han Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China;
| |
Collapse
|
9
|
Gatti DM, Tyler AL, Mahoney JM, Churchill GA, Yener B, Koyuncu D, Gurcan MN, Niazi M, Tavolara T, Gower AC, Dayao D, McGlone E, Ginese ML, Specht A, Alsharaydeh A, Tessier PA, Kurtz SL, Elkins K, Kramnik I, Beamer G. Systems genetics uncover new loci containing functional gene candidates in Mycobacterium tuberculosis-infected Diversity Outbred mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572738. [PMID: 38187647 PMCID: PMC10769337 DOI: 10.1101/2023.12.21.572738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Mycobacterium tuberculosis, the bacillus that causes tuberculosis (TB), infects 2 billion people across the globe, and results in 8-9 million new TB cases and 1-1.5 million deaths each year. Most patients have no known genetic basis that predisposes them to disease. We investigated the complex genetic basis of pulmonary TB by modelling human genetic diversity with the Diversity Outbred mouse population. When infected with M. tuberculosis, one-third develop early onset, rapidly progressive, necrotizing granulomas and succumb within 60 days. The remaining develop non-necrotizing granulomas and survive longer than 60 days. Genetic mapping using clinical indicators of disease, granuloma histopathological features, and immune response traits identified five new loci on mouse chromosomes 1, 2, 4, 16 and three previously identified loci on chromosomes 3 and 17. Quantitative trait loci (QTLs) on chromosomes 1, 16, and 17, associated with multiple correlated traits and had similar patterns of allele effects, suggesting these QTLs contain important genetic regulators of responses to M. tuberculosis. To narrow the list of candidate genes in QTLs, we used a machine learning strategy that integrated gene expression signatures from lungs of M. tuberculosis-infected Diversity Outbred mice with gene interaction networks, generating functional scores. The scores were then used to rank candidates for each mapped trait in each locus, resulting in 11 candidates: Ncf2, Fam20b, S100a8, S100a9, Itgb5, Fstl1, Zbtb20, Ddr1, Ier3, Vegfa, and Zfp318. Importantly, all 11 candidates have roles in infection, inflammation, cell migration, extracellular matrix remodeling, or intracellular signaling. Further, all candidates contain single nucleotide polymorphisms (SNPs), and some but not all SNPs were predicted to have deleterious consequences on protein functions. Multiple methods were used for validation including (i) a statistical method that showed Diversity Outbred mice carrying PWH/PhJ alleles on chromosome 17 QTL have shorter survival; (ii) quantification of S100A8 protein levels, confirming predicted allele effects; and (iii) infection of C57BL/6 mice deficient for the S100a8 gene. Overall, this work demonstrates that systems genetics using Diversity Outbred mice can identify new (and known) QTLs and new functionally relevant gene candidates that may be major regulators of granuloma necrosis and acute inflammation in pulmonary TB.
Collapse
Affiliation(s)
- D M Gatti
- The Jackson Laboratory, Bar Harbor, ME
| | - A L Tyler
- The Jackson Laboratory, Bar Harbor, ME
| | | | | | - B Yener
- Rensselaer Polytechnic Institute, Troy, NY
| | - D Koyuncu
- Rensselaer Polytechnic Institute, Troy, NY
| | - M N Gurcan
- Wake Forest University School of Medicine, Winston Salem, NC
| | - Mkk Niazi
- Wake Forest University School of Medicine, Winston Salem, NC
| | - T Tavolara
- Wake Forest University School of Medicine, Winston Salem, NC
| | - A C Gower
- Clinical and Translational Science Institute, Boston University, Boston, MA
| | - D Dayao
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA
| | - E McGlone
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA
| | - M L Ginese
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA
| | - A Specht
- Tufts University Cummings School of Veterinary Medicine, North Grafton, MA
| | - A Alsharaydeh
- Texas Biomedical Research Institute, San Antonio, TX
| | - P A Tessier
- Department of Microbiology and Immunology, Laval University School of Medicine, Quebec, Canada
| | - S L Kurtz
- Center for Biologics, Food and Drug Administration, Bethesda, MD
| | - K Elkins
- Center for Biologics, Food and Drug Administration, Bethesda, MD
| | - I Kramnik
- NIEDL, Boston University, Boston, MA
| | - G Beamer
- Texas Biomedical Research Institute, San Antonio, TX
| |
Collapse
|
10
|
Stoyanov D, Stoyanov GS, Ivanov MN, Spasov RH, Tonchev AB. Transcription Factor Zbtb20 as a Regulator of Malignancy and Its Practical Applications. Int J Mol Sci 2023; 24:13763. [PMID: 37762065 PMCID: PMC10530547 DOI: 10.3390/ijms241813763] [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: 08/03/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Zbtb20 (zinc finger and BTB domain-containing protein 20) is a transcription factor with a zinc finger DNA binding domain and a BTB domain responsible for protein-protein interaction. Recently, this TF has received attention because new data showed its pivotal involvement in normal neural development and its regulatory effects on proliferation and differentiation in different tissues. Zbtb20 was shown to increase proliferation and migration and confer resistance to apoptosis in the contexts of many malignant tumors like hepatocellular carcinoma, non-small-cell lung carcinoma, gastric adenocarcinoma, glioblastoma multiforme, breast cancer, and acute myeloid leukemia. The involvement of Zbtb20 in tumor biology is best studied in hepatocellular carcinoma, where it is a promising candidate as an immunohistochemical tumor marker or may be used in patient screening. Here we review the current data connecting Zbtb20 with malignant tumors.
Collapse
Affiliation(s)
- Dimo Stoyanov
- Department of Anatomy and Cell Biology, Medical University of Varna, 9000 Varna, Bulgaria
| | - George S. Stoyanov
- Department of Clinical Pathology, Complex Oncology Center, 9700 Shumen, Bulgaria
| | - Martin N. Ivanov
- Department of Anatomy and Cell Biology, Medical University of Varna, 9000 Varna, Bulgaria
- Department of Stem Cell Biology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
| | - Radoslav H. Spasov
- Department of Anatomy and Cell Biology, Medical University of Varna, 9000 Varna, Bulgaria
| | - Anton B. Tonchev
- Department of Anatomy and Cell Biology, Medical University of Varna, 9000 Varna, Bulgaria
- Department of Stem Cell Biology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
| |
Collapse
|
11
|
Preiss NK, Kamal Y, Wilkins OM, Li C, Kolling FW, Trask HW, Usherwood YK, Cheng C, Frost HR, Usherwood EJ. Characterizing control of memory CD8 T cell differentiation by BTB-ZF transcription factor Zbtb20. Life Sci Alliance 2023; 6:e202201683. [PMID: 37414528 PMCID: PMC10326419 DOI: 10.26508/lsa.202201683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/08/2023] Open
Abstract
Members of the BTB-ZF transcription factor family regulate the immune system. Our laboratory identified that family member Zbtb20 contributes to the differentiation, recall responses, and metabolism of CD8 T cells. Here, we report a characterization of the transcriptional and epigenetic signatures controlled by Zbtb20 at single-cell resolution during the effector and memory phases of the CD8 T cell response. Without Zbtb20, transcriptional programs associated with memory CD8 T cell formation were up-regulated throughout the CD8 T response. A signature of open chromatin was associated with genes controlling T cell activation, consistent with the known impact on differentiation. In addition, memory CD8 T cells lacking Zbtb20 were characterized by open chromatin regions with overrepresentation of AP-1 transcription factor motifs and elevated RNA- and protein-level expressions of the corresponding AP-1 components. Finally, we describe motifs and genomic annotations from the DNA targets of Zbtb20 in CD8 T cells identified by cleavage under targets and release under nuclease (CUT&RUN). Together, these data establish the transcriptional and epigenetic networks contributing to the control of CD8 T cell responses by Zbtb20.
Collapse
Affiliation(s)
- Nicholas K Preiss
- Microbiology and Immunology Department, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Yasmin Kamal
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Owen M Wilkins
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
- Genomics and Molecular Biology Shared Resource, Dartmouth Cancer Center, Geisel School of Medicine, Lebanon, NH, USA
| | - Chenyang Li
- Genomic Medicine Department, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center UTHealth Houston, Houston, TX, USA
| | - Fred W Kolling
- Genomics and Molecular Biology Shared Resource, Dartmouth Cancer Center, Geisel School of Medicine, Lebanon, NH, USA
| | - Heidi W Trask
- Genomics and Molecular Biology Shared Resource, Dartmouth Cancer Center, Geisel School of Medicine, Lebanon, NH, USA
| | - Young-Kwang Usherwood
- Microbiology and Immunology Department, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Hildreth R Frost
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Edward J Usherwood
- Microbiology and Immunology Department, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
12
|
Tian Y, Ma X, Jiang Y, Han J, Zhang R, Xu X, Zhang W, Man C. Circular RNA circAKIRIN2 participates in the process of stress-induced immunosuppression affecting immune response to infectious bursal disease virus vaccine in chicken. Vet Microbiol 2023; 281:109746. [PMID: 37075663 DOI: 10.1016/j.vetmic.2023.109746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/10/2023] [Accepted: 04/15/2023] [Indexed: 04/21/2023]
Abstract
At present, stress-induced immunosuppression is still a hidden threat that leads to immunization failure and outbreaks of poultry diseases, and causes huge economic losses to the modern poultry industry. However, the molecular mechanisms of stress-induced immunosuppression affecting viral vaccine immunity are still poorly understood. Here, we identified circAKIRIN2 as a conserved circular transcript in chicken, and explored its expression patterns in different immune states by quantitative real-time PCR (qRT-PCR), then conducted bioinformatics analysis. The results showed that circAKIRIN2 actively participated in the process of stress-induced immunosuppression affecting the immune response to infectious bursal disease virus (IBDV) vaccine. The key time points for circAKIRIN2 involving in the process were 2 day post immunization (dpi), 5 dpi, and 28 dpi, especially at the acquired immune stage. The important tissues that responded to the process included the heart, liver, and lung, all of which changed significantly. In addition, circAKIRIN2 as a competing endogenous RNA (ceRNA) sponging zinc finger and BTB domain containing 20 (ZBTB20) was a potential molecular mechanism for regulating immune functions in the process. In conclusion, circAKIRIN2 is a key regulatory factor for stress-induced immunosuppression affecting the IBDV vaccine immune response, and this study can provide a new perspective for exploring the molecular regulatory mechanisms of stress-induced immunosuppression affecting immune response.
Collapse
Affiliation(s)
- Yufei Tian
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China
| | - Xiaoli Ma
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China
| | - Yi Jiang
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China
| | - Jianwei Han
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China
| | - Rui Zhang
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China
| | - Xinxin Xu
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China
| | - Wei Zhang
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China
| | - Chaolai Man
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China.
| |
Collapse
|
13
|
Wang Y, Cai M, Lou Y, Zhang S, Liu X. ZBTB20-AS1 promoted Alzheimer's disease progression through ZBTB20/GSK-3β/Tau pathway. Biochem Biophys Res Commun 2023; 640:88-96. [PMID: 36502636 DOI: 10.1016/j.bbrc.2022.11.107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/30/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022]
Abstract
To elucidate the potential molecular mechanisms of ZBTB20-AS1 on ZBTB20 and GSK-3β/Tau signaling pathway in the pathogenesis of Alzheimer's disease (AD), SH-SY5Y cells were obtained for in vitro experiments and AD models were constructed using β-Amyloid 1-42. CCK8 assay was implemented for determining cell viability. Flow cytometry was used for cell apoptosis detection. Dual-luciferase reporter and RNA-RNA pull down assay was employed for elucidating molecular interactions. Immunohistochemistry, RT-qPCR and western blotting were performed for measuring gene expression. The results showed that expression of LncRNA ZBTB20-AS1 was significantly upregulated, while ZBTB20 was downregulated in SH-SY5Y-AD cells. ZBTB20 was the target gene of LncRNA ZBTB20-AS1. Overexpression of ZBTB20 or knockdown of LncRNA ZBTB20-AS1 inhibited SH-SY5Y-AD cells apoptosis and suppressed GSK3β/Tau pathway, and knockdown of ZBTB20-AS1 increased cell viability and decreased apoptosis. In conclusion, overexpression of ZBTB20-AS1 inhibited ZBTB20 expression and promoted GSK-3β expression and Tau phosphorylation, contributing to the development of AD.
Collapse
Affiliation(s)
- Yanwen Wang
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Zhejiang, 310013, China
| | - Miao Cai
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Zhejiang, 310013, China
| | - Yue Lou
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Zhejiang, 310013, China
| | - Siran Zhang
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Zhejiang, 310013, China
| | - Xiaoli Liu
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Zhejiang, 310013, China.
| |
Collapse
|
14
|
Lu L, Shi M, Qiu J, Shi Z, Wang C, Fu Y, Lin C, Zhang L, Tao J, Liu C, Wei F, Liang S, Zheng J. ZBTB20 regulates cardiac allograft rejection through NFкB-mediated inflammation in mouse heart transplantation. Transpl Immunol 2022; 74:101676. [PMID: 35872083 DOI: 10.1016/j.trim.2022.101676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022]
Abstract
Allograft rejection is a major obstacle for the long-term survival of heart transplantation (Htx) patients. The cardiac allograft rejection requires the activation of macrophages and effector T cells. In this study, we explored the role of zinc-finger and BTB domain containing protein 20 (ZBTB20) in the regulation of heart allograft rejection. Flow cytometry analysis of the spleen cells from mice undergoing an acute cardiac rejection revealed that the ZBTB20 protein expression was upregulated in both T and B cells(n = 4,P < 0.01). In addition, ZBTB20 gene knockdown significantly prolonged the survival of heart allografts in mice(n = 4,P < 0.01). Lack of ZBTB20 increased the expression of Foxp3 and limited the response of T helper 1 (Th1) cells(n = 4,P < 0.01). The ZBTB20-related regulation occurred through the activation of the NFкB pathway. In conclusion, our data suggest that ZBTB20 is involved in the regulation of T cells involved in acute heart allograft rejection. Hence, downregulation of ZBTB20 expression may inhibit T cells to prolong heart transplant survival.
Collapse
Affiliation(s)
- Liuyi Lu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macao SAR, China
| | - Maomao Shi
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Junxiong Qiu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhanyue Shi
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Chao Wang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Thoracic and Cardiac Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Yuan Fu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Canxiang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou 510630, China
| | - Lisui Zhang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jun Tao
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Chang Liu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Feng Wei
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shi Liang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Junmeng Zheng
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Surgery, Kiang Wu Hospital, Macao SAR, China.
| |
Collapse
|
15
|
Krzyzanowska AK, Haynes Ii RAH, Kovalovsky D, Lin HC, Osorio L, Edelblum KL, Corcoran LM, Rabson AB, Denzin LK, Sant'Angelo DB. Zbtb20 identifies and controls a thymus-derived population of regulatory T cells that play a role in intestinal homeostasis. Sci Immunol 2022; 7:eabf3717. [PMID: 35522722 DOI: 10.1126/sciimmunol.abf3717] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The expression of BTB-ZF transcription factors such as ThPOK in CD4+ T cells, Bcl6 in T follicular helper cells, and PLZF in natural killer T cells defines the fundamental nature and characteristics of these cells. Screening for lineage-defining BTB-ZF genes led to the discovery of a subset of T cells that expressed Zbtb20. About half of Zbtb20+ T cells expressed FoxP3, the lineage-defining transcription factor for regulatory T cells (Tregs). Zbtb20+ Tregs were phenotypically and genetically distinct from the larger conventional Treg population. Zbtb20+ Tregs constitutively expressed mRNA for interleukin-10 and produced high levels of the cytokine upon primary activation. Zbtb20+ Tregs were enriched in the intestine and specifically expanded when inflammation was induced by the use of dextran sodium sulfate. Conditional deletion of Zbtb20 in T cells resulted in a loss of intestinal epithelial barrier integrity. Consequently, knockout (KO) mice were acutely sensitive to colitis and often died because of the disease. Adoptive transfer of Zbtb20+ Tregs protected the Zbtb20 conditional KO mice from severe colitis and death, whereas non-Zbtb20 Tregs did not. Zbtb20 was detected in CD24hi double-positive and CD62Llo CD4 single-positive thymocytes, suggesting that expression of the transcription factor and the phenotype of these cells were induced during thymic development. However, Zbtb20 expression was not induced in "conventional" Tregs by activation in vitro or in vivo. Thus, Zbtb20 expression identified and controlled the function of a distinct subset of Tregs that are involved in intestinal homeostasis.
Collapse
Affiliation(s)
- Agata K Krzyzanowska
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.,Rutgers Graduate School of Biomedical Sciences, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Rashade A H Haynes Ii
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Damian Kovalovsky
- Center for Cancer Research, National Cancer Institute National Institutes of Health, Bethesda, MD 20892, USA
| | - Hsin-Ching Lin
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Louis Osorio
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Karen L Edelblum
- Department of Pathology and Laboratory Medicine Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Lynn M Corcoran
- The Walter and Eliza Hall Institute of Medical Research Immunology Division, Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Arnold B Rabson
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.,Rutgers Graduate School of Biomedical Sciences, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.,Department of Pediatrics and Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Lisa K Denzin
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.,Rutgers Graduate School of Biomedical Sciences, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.,Department of Pediatrics and Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Derek B Sant'Angelo
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.,Rutgers Graduate School of Biomedical Sciences, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.,Department of Pediatrics and Pharmacology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| |
Collapse
|
16
|
Jin Y, Li Z, Wu Y, Li H, Liu Z, Liu L, Ouyang N, Zhou T, Fang B, Xia L. Aberrant Fluid Shear Stress Contributes to Articular Cartilage Pathogenesis via Epigenetic Regulation of ZBTB20 by H3K4me3. J Inflamm Res 2021; 14:6067-6083. [PMID: 34824542 PMCID: PMC8610757 DOI: 10.2147/jir.s339382] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/05/2021] [Indexed: 01/21/2023] Open
Abstract
Purpose Osteoarthritis (OA) is a common disease for human beings, characterized by severe inflammation, cartilage degradation, and subchondral bone destruction. However, current therapies are limited to relieving pain or joint replacement and no effective treatment methods have been discovered to improve degenerative changes. Currently, a variety of evidences have indicated that aberrant mechanical stimuli is closely associated with articular joint pathogenesis, while the detailed underlying mechanism remains unelucidated. In the present study, we determined to investigate the impact of excessive high fluid shear stress (FSS) on primary chondrocytes and the underlying epigenetic mechanisms. Materials and Methods Phalloidin staining and EdU staining were used to evaluate cell morphology and viability. The mRNA level and protein level of genes were determined by qPCR, Western blot assay, and immunofluorescence staining. Mechanistic investigation was performed through RNA-sequencing and CUT&Tag sequencing. In vivo, we adopted unilateral anterior crossbites (UAC) mice model to investigate the expression of H3K4me3 and ZBTB20 in aberrant force-related cartilage pathogenesis. Results The results demonstrated that FSS greatly disrupts cell morphology and significantly decreased chondrocyte viability. Aberrant FSS induces remarkable inflammatory mediators production, leading to cartilage degeneration and degradation. In depth mechanistic study showed that FSS results in more than 10-fold upregulation of H3K4me3, and the modulatory effect of H3K4me3 on cartilage was obtained by directly targeting ZBTB20. Furthermore, Wnt signaling was strongly activated in high FSS-induced OA pathogenesis, and the negative impact of ZBTB20 on chondrocytes was also achieved through activating Wnt signaling pathway. Moreover, pharmacological inhibition of H3K4me3 activation by MM-102 or treatment with Wnt pathway inhibitor LF3 could effectively alleviate the destructive effect of FSS on chondrocytes. In vivo UAC mice model validated the dysregulation of H3K4me3 and ZBTB20 in aberrant force-induced cartilage pathogenesis. Conclusion Through the combination of in vitro FSS model and in vivo UAC model, KMT2B-H3K4me3-ZBTB20 axis was first identified in aberrant FSS-induced cartilage pathogenesis, which may provide evidences for epigenetic-based therapy in the future.
Collapse
Affiliation(s)
- Yu Jin
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, People's Republic of China
| | - Zhenxia Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, People's Republic of China
| | - Yanran Wu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, People's Republic of China
| | - Hairui Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, People's Republic of China
| | - Zhen Liu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, People's Republic of China
| | - Lu Liu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, People's Republic of China
| | - Ningjuan Ouyang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, People's Republic of China
| | - Ting Zhou
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, People's Republic of China
| | - Bing Fang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, People's Republic of China
| | - Lunguo Xia
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, People's Republic of China
| |
Collapse
|
17
|
Cheng ZY, He TT, Gao XM, Zhao Y, Wang J. ZBTB Transcription Factors: Key Regulators of the Development, Differentiation and Effector Function of T Cells. Front Immunol 2021; 12:713294. [PMID: 34349770 PMCID: PMC8326903 DOI: 10.3389/fimmu.2021.713294] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
The development and differentiation of T cells represents a long and highly coordinated, yet flexible at some points, pathway, along which the sequential and dynamic expressions of different transcriptional factors play prominent roles at multiple steps. The large ZBTB family comprises a diverse group of transcriptional factors, and many of them have emerged as critical factors that regulate the lineage commitment, differentiation and effector function of hematopoietic-derived cells as well as a variety of other developmental events. Within the T-cell lineage, several ZBTB proteins, including ZBTB1, ZBTB17, ZBTB7B (THPOK) and BCL6 (ZBTB27), mainly regulate the development and/or differentiation of conventional CD4/CD8 αβ+ T cells, whereas ZBTB16 (PLZF) is essential for the development and function of innate-like unconventional γδ+ T & invariant NKT cells. Given the critical role of T cells in host defenses against infections/tumors and in the pathogenesis of many inflammatory disorders, we herein summarize the roles of fourteen ZBTB family members in the development, differentiation and effector function of both conventional and unconventional T cells as well as the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Zhong-Yan Cheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ting-Ting He
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiao-Ming Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Ying Zhao
- Department of Pathophysiology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Jun Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
18
|
Li F, Du M, Yang Y, Wang Z, Zhang H, Wang X, Li Q. Zinc finger and BTB domain-containing protein 20 aggravates angiotensin II-induced cardiac remodeling via the EGFR-AKT pathway. J Mol Med (Berl) 2021; 100:427-438. [PMID: 34232352 DOI: 10.1007/s00109-021-02103-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/05/2021] [Accepted: 06/10/2021] [Indexed: 11/26/2022]
Abstract
Zinc finger and BTB domain-containing protein 20 (ZBTB20) play an important role in glucose and lipid homeostasis. ZBTB20 was shown to be a crucial protein for the maintenance of cardiac contractile function. However, the role of ZBTB20 in cardiac response remodeling has not been elucidated. Thus, this study aimed to explore the role of ZBTB20 in cardiac remodeling following angiotensin II insult. Mice were subjected to angiotensin II infusion to induce a cardiac adverse remodeling model. An adeno-associated virus (AAV) 9 system was used to deliver ZBTB20 to the mouse heart. Here, we demonstrate that ZBTB20 expression is elevated in angiotensin II-induced cardiac remodeling and in response to cardiomyocyte insults. Furthermore, AAV9-mediated overexpression of ZBTB20 caused cardiac wall hypertrophy, chamber dilation, increased fibrosis, and reduced ejection fraction. Additionally, ZBTB20 siRNA protected cardiomyocytes from angiotensin II-induced hypertrophy. Mechanistically, ZBTB20 interferes with EGFR and Akt signaling and modulates the remodeling response. Overexpression of constitutively active Akt counteracts ZBTB20 knockdown-mediated protection of adverse cardiac remodeling. These findings illustrate the role of ZBTB20 in the transition of adverse cardiac remodeling toward heart failure and provide evidence for the molecular programs inducing adverse cardiac remodeling. KEY MESSAGES: ZBTB20 is a transcription factor from the POK family. ZBTB20 is upregulated in heart tissue treated with angiotensin II. ZBTB20 influences cardiomyocyte hypertrophy via the EGFR-Akt pathway. Akt continuous activation leads to similar results to ZBTB20 overexpression.
Collapse
Affiliation(s)
- Fangfang Li
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221000, People's Republic of China
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, 221000, China
| | - Miaomiao Du
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221000, People's Republic of China
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, 221000, China
| | - Yiming Yang
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221000, People's Republic of China
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, 221000, China
| | - Zhu Wang
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221000, People's Republic of China
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, 221000, China
| | - Hu Zhang
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221000, People's Republic of China
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, 221000, China
| | - Xiaoyu Wang
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221000, People's Republic of China
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, 221000, China
| | - Qing Li
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, 221000, People's Republic of China.
- Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, 221000, China.
| |
Collapse
|
19
|
ZBTB20 Positively Regulates Oxidative Stress, Mitochondrial Fission, and Inflammatory Responses of ox-LDL-Induced Macrophages in Atherosclerosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5590855. [PMID: 33777314 PMCID: PMC7972849 DOI: 10.1155/2021/5590855] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/26/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023]
Abstract
Atherosclerosis (AS) is one of the most serious and common cardiovascular diseases affecting human health. AS is featured by the accumulation of plaques in vessel walls. The pathophysiology of AS is relevant in the low-density lipoprotein (LDL) uptake by macrophages, as well as the conversion of macrophages to foam cells. However, the mechanisms about how macrophages regulate AS have not been fully elucidated. In this study, we aimed to illuminate the roles of ZBTB20 and to excavate the underlying regulative mechanisms of ZBTB20 in AS. The microarray analysis revealed that ZBTB20 was a hub gene in the oxidative stress and inflammatory responses induced by oxidized LDL (ox-LDL) in AS. Correspondingly, our validation studies showed that ZBTB20 increased in either the human atherosclerotic lesion or the ox-LDL-stimulated macrophages. Moreover, the knockdown of ZBTB20 decreased M1 polarization, suppressed the proinflammatory factors, inhibited mitochondrial fission, and reduced the oxidative stress level of macrophages induced by ox-LDL. The mechanistic studies revealed that the ZBTB20 knockdown suppressed NF-κB/MAPK activation and attenuated the mitochondrial fission possibly via regulating the nucleus translocation of NRF2, a pivotal transcription factor on redox homeostasis. Our in vivo studies showed that the sh-ZBTB20 adenovirus injection could reduce the progression of AS in apolipoprotein E-deficient (ApoE−/−) mice. All in all, these results suggested that ZBTB20 positively regulated the oxidative stress level, mitochondrial fission, and inflammatory responses of macrophages induced by ox-LDL, and the knockdown of ZBTB20 could attenuate the development of AS in ApoE−/− mice.
Collapse
|