1
|
Xu P, Xiao Y, Xiao Z, Li J. Developing a genome-wide long sequence-specific tag for sex identification in spotted knifejaw (Oplegnathus punctatus). Mol Genet Genomics 2025; 300:32. [PMID: 40106105 DOI: 10.1007/s00438-025-02240-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Spotted knifejaw (Oplegnathus punctatus), an economically important species in marine aquaculture, employs a unique sex determination mechanism based on a complex sex chromosome system (X1X1X2X2/X1X2Y). Males (2n = 47) possess one fewer chromosome than females (2n = 48), and their karyotype includes an unusually large neo-Y chromosome. Additionally, a pronounced sexual dimorphism in growth rate is observed, with males exhibiting a faster growth rate than females. In this study, we conducted a comprehensive whole-genome scan, which initially revealed structural variations in the anti-inflammatory itih4 gene between male and female O. punctatus. Additionally, we designed a pair of primers to detect DNA sequence variations within the itih4a/itih4b gene. These variations are located in the intergenic region of the fusion Y chromosome in male O. punctatus, compared to the homologous X chromosome in females. In females without DNA insertions in the itih4a/itih4b intergenic region, a single band of 351 bp is amplified. By contrast, in males with DNA insertions, two bands are amplified (755 bp and 351 bp). The 755 bp band specifically indicates the presence of a DNA insertion in the itih4a/itih4b intergenic region on the Y chromosome, associated with male-specific genetic traits. Our study will facilitate the rapid identification of the genetic sex of both male and female O. punctatus individuals.
Collapse
Affiliation(s)
- Pingrui Xu
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Qingdao, China
| | - Yongshuang Xiao
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Qingdao, China.
| | - Zhizhong Xiao
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Qingdao, China
| | - Jun Li
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Qingdao, China.
| |
Collapse
|
2
|
Zeng Y, Buonfiglio F, Li J, Pfeiffer N, Gericke A. Mechanisms Underlying Vascular Inflammaging: Current Insights and Potential Treatment Approaches. Aging Dis 2025:AD.2024.0922. [PMID: 39812546 DOI: 10.14336/ad.2024.0922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/18/2024] [Indexed: 01/16/2025] Open
Abstract
Inflammaging refers to chronic, low-grade inflammation that becomes more common with age and plays a central role in the pathophysiology of various vascular diseases. Key inflammatory mediators involved in inflammaging contribute to endothelial dysfunction and accelerate the progression of atherosclerosis. In addition, specific pathological mechanisms and the role of inflammasomes have emerged as critical drivers of immune responses within the vasculature. A comprehensive understanding of these processes may lead to innovative treatment strategies that could significantly improve the management of age-related vascular diseases. Emerging therapeutic approaches, including cytokine inhibitors, senolytics, and specialized pro-resolving mediators, aim to counteract inflammaging and restore vascular health. This review seeks to provide an in-depth exploration of the molecular pathways underlying vascular inflammaging and highlight potential therapeutic interventions.
Collapse
|
3
|
Bhat MF, Srdanović S, Sundberg LR, Einarsdóttir HK, Marjomäki V, Dekker FJ. Impact of HDAC inhibitors on macrophage polarization to enhance innate immunity against infections. Drug Discov Today 2024; 29:104193. [PMID: 39332483 DOI: 10.1016/j.drudis.2024.104193] [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: 05/31/2024] [Revised: 08/26/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Innate immunity plays an important role in host defense against pathogenic infections. It involves macrophage polarization into either the pro-inflammatory M1 or the anti-inflammatory M2 phenotype, influencing immune stimulation or suppression, respectively. Epigenetic changes during immune reactions contribute to long-term innate immunity imprinting on macrophage polarization. It is becoming increasingly evident that epigenetic modulators, such as histone deacetylase (HDAC) inhibitors (HDACi), enable the enhancement of innate immunity by tailoring macrophage polarization in response to immune stressors. In this review, we summarize current literature on the impact of HDACi and other epigenetic modulators on the functioning of macrophages during diseases that have a strong immune component, such as infections. Depending on the disease context and the chosen therapeutic intervention, HDAC1, HDAC2, HDAC3, HDAC6, or HDAC8 are particularly important in influencing macrophage polarization towards either M1 or M2 phenotypes. We anticipate that therapeutic strategies based on HDAC epigenetic mechanisms will provide a unique approach to boost immunity against disease challenges, including resistant infections.
Collapse
Affiliation(s)
- Mohammad Faizan Bhat
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | - Sonja Srdanović
- Akthelia Pharmaceuticals, Grandagardi 16, 101 Reykjavik, Iceland
| | - Lotta-Riina Sundberg
- Department of Biological and Environmental Sciences and Nanoscience Center, 40014 University of Jyväskylä, Jyväskylä, Finland
| | | | - Varpu Marjomäki
- Department of Biological and Environmental Sciences and Nanoscience Center, 40014 University of Jyväskylä, Jyväskylä, Finland
| | - Frank J Dekker
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
4
|
Halasz H, Malekos E, Covarrubias S, Yitiz S, Montano C, Sudek L, Katzman S, Liu SJ, Horlbeck MA, Namvar L, Weissman JS, Carpenter S. CRISPRi screens identify the lncRNA, LOUP, as a multifunctional locus regulating macrophage differentiation and inflammatory signaling. Proc Natl Acad Sci U S A 2024; 121:e2322524121. [PMID: 38781216 PMCID: PMC11145268 DOI: 10.1073/pnas.2322524121] [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: 12/20/2023] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) account for the largest portion of RNA from the transcriptome, yet most of their functions remain unknown. Here, we performed two independent high-throughput CRISPRi screens to understand the role of lncRNAs in monocyte function and differentiation. The first was a reporter-based screen to identify lncRNAs that regulate TLR4-NFkB signaling in human monocytes and the second screen identified lncRNAs involved in monocyte to macrophage differentiation. We successfully identified numerous noncoding and protein-coding genes that can positively or negatively regulate inflammation and differentiation. To understand the functional roles of lncRNAs in both processes, we chose to further study the lncRNA LOUP [lncRNA originating from upstream regulatory element of SPI1 (also known as PU.1)], as it emerged as a top hit in both screens. Not only does LOUP regulate its neighboring gene, the myeloid fate-determining factor SPI1, thereby affecting monocyte to macrophage differentiation, but knockdown of LOUP leads to a broad upregulation of NFkB-targeted genes at baseline and upon TLR4-NFkB activation. LOUP also harbors three small open reading frames capable of being translated and are responsible for LOUP's ability to negatively regulate TLR4/NFkB signaling. This work emphasizes the value of high-throughput screening to rapidly identify functional lncRNAs in the innate immune system.
Collapse
Affiliation(s)
- Haley Halasz
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA95064
| | - Eric Malekos
- Department of Biomolecular Engineering, University of California Santa Cruz, CA95064
| | - Sergio Covarrubias
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA95064
| | - Samira Yitiz
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA95064
| | - Christy Montano
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA95064
| | - Lisa Sudek
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA95064
| | - Sol Katzman
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA95064
| | - S. John Liu
- Department of Radiation Oncology, University of California, San Francisco, CA94158
- Department of Neurological Surgery, University of California, San Francisco, CA94158
| | - Max A. Horlbeck
- Department of Radiation Oncology, University of California, San Francisco, CA94158
- Department of Neurological Surgery, University of California, San Francisco, CA94158
- Department of Pediatrics, Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA02115
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA02138
| | - Leila Namvar
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA95064
| | - Jonathan S. Weissman
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA02142
- HHMI, Chevy Chase, MD20815
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02142
| | - Susan Carpenter
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, CA95064
| |
Collapse
|
5
|
Karpurapu M, Kakarala KK, Chung S, Nie Y, Koley A, Dougherty P, Christman JW. Epigallocatechin gallate regulates the myeloid-specific transcription factor PU.1 in macrophages. PLoS One 2024; 19:e0301904. [PMID: 38662666 PMCID: PMC11045095 DOI: 10.1371/journal.pone.0301904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Our previous research demonstrated that PU.1 regulates expression of the genes involved in inflammation in macrophages. Selective knockdown of PU.1 in macrophages ameliorated LPS-induced acute lung injury (ALI) in bone marrow chimera mice. Inhibitors that block the transcriptional activity of PU.1 in macrophages have the potential to mitigate the pathophysiology of LPS-induced ALI. However, complete inactivation of PU.1 gene disrupts normal myelopoiesis. Although the green tea polyphenol Epigallocatechin gallate (EGCG) has been shown to regulate inflammatory genes in various cell types, it is not known if EGCG alters the transcriptional activity of PU.1 protein. Using Schrodinger Glide docking, we have identified that EGCG binds with PU.1 protein, altering its DNA-binding and self-dimerization activity. In silico analysis shows that EGCG forms Hydrogen bonds with Glutamic Acid 209, Leucine 250 in DNA binding and Lysine 196, Tryptophan 193, and Leucine 182 in the self-dimerization domain of the PU.1 protein. Experimental validation using mouse bone marrow-derived macrophages (BMDM) confirmed that EGCG inhibits both DNA binding by PU.1 and self-dimerization. Importantly, EGCG had no impact on expression of the total PU.1 protein levels but significantly reduced expression of various inflammatory genes and generation of ROS. In summary, we report that EGCG acts as an inhibitor of the PU.1 transcription factor in macrophages.
Collapse
Affiliation(s)
- Manjula Karpurapu
- Division of Pulmonary, Davis Heart and Lung Research Institute, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | | | - Sangwoon Chung
- Division of Pulmonary, Davis Heart and Lung Research Institute, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| | - Yunjuan Nie
- Division of Pulmonary, Davis Heart and Lung Research Institute, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Columbus, OH, United States of America
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, P.R. China
| | - Amritendu Koley
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States of America
| | - Patrick Dougherty
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States of America
| | - John W. Christman
- Division of Pulmonary, Davis Heart and Lung Research Institute, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Columbus, OH, United States of America
| |
Collapse
|
6
|
Ahmad A. Epigenetic regulation of inflammation. Semin Cell Dev Biol 2024; 154:165-166. [PMID: 37689497 DOI: 10.1016/j.semcdb.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2023]
Affiliation(s)
- Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
7
|
Deng JY, Wu XQ, He WJ, Liao X, Tang M, Nie XQ. Targeting DNA methylation and demethylation in diabetic foot ulcers. J Adv Res 2023; 54:119-131. [PMID: 36706989 PMCID: PMC10703625 DOI: 10.1016/j.jare.2023.01.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Poor wound healing is a significant complication of diabetes, which is commonly caused by neuropathy, trauma, deformities, plantar hypertension and peripheral arterial disease. Diabetic foot ulcers (DFU) are difficult to heal, which makes patients susceptible to infections and can ultimately conduce to limb amputation or even death in severe cases. An increasing number of studies have found that epigenetic alterations are strongly associated with poor wound healing in diabetes. AIM OF REVIEW This work provides significant insights into the development of therapeutics for improving chronic diabetic wound healing, particularly by targeting and regulating DNA methylation and demethylation in DFU. Key scientific concepts of review: DNA methylation and demethylation play an important part in diabetic wound healing, via regulating corresponding signaling pathways in different breeds of cells, including macrophages, vascular endothelial cells and keratinocytes. In this review, we describe the four main phases of wound healing and their abnormality in diabetic patients. Furthermore, we provided an in-depth summary and discussion on how DNA methylation and demethylation regulate diabetic wound healing in different types of cells; and gave a brief summary on recent advances in applying cellular reprogramming techniques for improving diabetic wound healing.
Collapse
Affiliation(s)
- Jun-Yu Deng
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi 563006, China; Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Xing-Qian Wu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Wen-Jie He
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Xin Liao
- Affiliated Hospital of Zunyi Medical University, Zunyi 563006, China
| | - Ming Tang
- Queensland University of Technology (QUT), School of Biomedical Sciences, Centre for Genomics and Personalized Health at the Translational Research Institute (TRI), Brisbane, QLD 4102, Australia.
| | - Xu-Qiang Nie
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi 563006, China; Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Queensland University of Technology (QUT), School of Biomedical Sciences, Centre for Genomics and Personalized Health at the Translational Research Institute (TRI), Brisbane, QLD 4102, Australia.
| |
Collapse
|
8
|
Fu B, Xiong Y, Sha Z, Xue W, Xu B, Tan S, Guo D, Lin F, Wang L, Ji J, Luo Y, Lin X, Wu H. SEPTIN2 suppresses an IFN-γ-independent, proinflammatory macrophage activation pathway. Nat Commun 2023; 14:7441. [PMID: 37978190 PMCID: PMC10656488 DOI: 10.1038/s41467-023-43283-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
Interferon-gamma (IFN-γ) signaling is necessary for the proinflammatory activation of macrophages but IFN-γ-independent pathways, for which the initiating stimuli and downstream mechanisms are lesser known, also contribute. Here we identify, by high-content screening, SEPTIN2 (SEPT2) as a negative regulation of IFN-γ-independent macrophage autoactivation. Mechanistically, endoplasmic reticulum (ER) stress induces the expression of SEPT2, which balances the competition between acetylation and ubiquitination of heat shock protein 5 at position Lysine 327, thereby alleviating ER stress and constraining M1-like polarization and proinflammatory cytokine release. Disruption of this negative feedback regulation leads to the accumulation of unfolded proteins, resulting in accelerated M1-like polarization, excessive inflammation and tissue damage. Our study thus uncovers an IFN-γ-independent macrophage proinflammatory autoactivation pathway and suggests that SEPT2 may play a role in the prevention or resolution of inflammation during infection.
Collapse
Affiliation(s)
- Beibei Fu
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Zhou Sha
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing University, 401331, Chongqing, China
| | - Binbin Xu
- School of Pharmaceutical Sciences, Chongqing University, 401331, Chongqing, China
| | - Shun Tan
- Chongqing Public Health Medical Center, 400036, Chongqing, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Feng Lin
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Lulu Wang
- School of Life Sciences, Chongqing University, 401331, Chongqing, China
| | - Jianjian Ji
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, 210023, Nanjing, China
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing University, 400044, Chongqing, China.
| | - Xiaoyuan Lin
- Institut für Virologie, Freie Universität Berlin, Robert-von-Ostertag-Str. 7-13, 14163, Berlin, Germany.
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), 400038, Chongqing, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, 401331, Chongqing, China.
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing University, 400044, Chongqing, China.
| |
Collapse
|
9
|
Maity J, Majumder S, Pal R, Saha B, Mukhopadhyay PK. Ascorbic acid modulates immune responses through Jumonji-C domain containing histone demethylases and Ten eleven translocation (TET) methylcytosine dioxygenase. Bioessays 2023; 45:e2300035. [PMID: 37694689 DOI: 10.1002/bies.202300035] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/12/2023]
Abstract
Ascorbic acid is a redox regulator in many physiological processes. Besides its antioxidant activity, many intriguing functions of ascorbic acid in the expression of immunoregulatory genes have been suggested. Ascorbic acid acts as a co-factor for the Fe+2 -containing α-ketoglutarate-dependent Jumonji-C domain-containing histone demethylases (JHDM) and Ten eleven translocation (TET) methylcytosine dioxygenasemediated epigenetic modulation. By influencing JHDM and TET, ascorbic acid facilitates the differentiation of double negative (CD4- CD8- ) T cells to double positive (CD4+ CD8+ ) T cells and of T-helper cells to different effector subsets. Ascorbic acid modulates plasma cell differentiation and promotes early differentiation of hematopoietic stem cells (HSCs) to NK cells. These findings indicate that ascorbic acid plays a significant role in regulating both innate and adaptive immune cells, opening up new research areas in Immunonutrition. Being a water-soluble vitamin and a safe micro-nutrient, ascorbic acid can be used as an adjunct therapy for many disorders of the immune system.
Collapse
Affiliation(s)
- Jeet Maity
- Department of Life Sciences, Presidency University, Kolkata, India
| | | | - Ranjana Pal
- Department of Life Sciences, Presidency University, Kolkata, India
| | | | | |
Collapse
|
10
|
Russo M, Pileri F, Ghisletti S. Novel insights into the role of acetyl-CoA producing enzymes in epigenetic regulation. Front Endocrinol (Lausanne) 2023; 14:1272646. [PMID: 37842307 PMCID: PMC10570720 DOI: 10.3389/fendo.2023.1272646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
Inflammation-dependent changes in gene expression programs in innate immune cells, such as macrophages, involve extensive reprogramming of metabolism. This reprogramming is essential for the production of metabolites required for chromatin modifications, such as acetyl-CoA, and regulate their usage and availability impacting the macrophage epigenome. One of the most transcriptionally induced proinflammatory mediator is nitric oxide (NO), which has been shown to inhibit key metabolic enzymes involved in the production of these metabolites. Recent evidence indicates that NO inhibits mitochondrial enzymes such as pyruvate dehydrogenase (PDH) in macrophages induced by inflammatory stimulus. PDH is involved in the production of acetyl-CoA, which is essential for chromatin modifications in the nucleus, such as histone acetylation. In addition, acetyl-CoA levels in inflamed macrophages are regulated by ATP citrate lyase (ACLY) and citrate transporter SLC25A1. Interestingly, acetyl-CoA producing enzymes, such as PDH and ACLY, have also been reported to be present in the nucleus and to support the local generation of cofactors such as acetyl-CoA. Here, we will discuss the mechanisms involved in the regulation of acetyl-CoA production by metabolic enzymes, their inhibition by prolonged exposure to inflammation stimuli, their involvement in dynamic inflammatory expression changes and how these emerging findings could have significant implications for the design of novel therapeutic approaches.
Collapse
Affiliation(s)
| | | | - Serena Ghisletti
- Department of Experimental Oncology, European Institute of Oncology (IEO) IRCCS, Milan, Italy
| |
Collapse
|
11
|
Jeong R, Bulyk ML. Blood cell traits' GWAS loci colocalization with variation in PU.1 genomic occupancy prioritizes causal noncoding regulatory variants. CELL GENOMICS 2023; 3:100327. [PMID: 37492098 PMCID: PMC10363807 DOI: 10.1016/j.xgen.2023.100327] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/10/2023] [Accepted: 04/25/2023] [Indexed: 07/27/2023]
Abstract
Genome-wide association studies (GWASs) have uncovered numerous trait-associated loci across the human genome, most of which are located in noncoding regions, making interpretation difficult. Moreover, causal variants are hard to statistically fine-map at many loci because of widespread linkage disequilibrium. To address this challenge, we present a strategy utilizing transcription factor (TF) binding quantitative trait loci (bQTLs) for colocalization analysis to identify trait associations likely mediated by TF occupancy variation and to pinpoint likely causal variants using motif scores. We applied this approach to PU.1 bQTLs in lymphoblastoid cell lines and blood cell trait GWAS data. Colocalization analysis revealed 69 blood cell trait GWAS loci putatively driven by PU.1 occupancy variation. We nominate PU.1 motif-altering variants as the likely shared causal variants at 51 loci. Such integration of TF bQTL data with other GWAS data may reveal transcriptional regulatory mechanisms and causal noncoding variants underlying additional complex traits.
Collapse
Affiliation(s)
- Raehoon Jeong
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Bioinformatics and Integrative Genomics Graduate Program, Harvard University, Cambridge, MA 02138, USA
| | - Martha L. Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Bioinformatics and Integrative Genomics Graduate Program, Harvard University, Cambridge, MA 02138, USA
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
12
|
Karnam K, Sedmaki K, Sharma P, Mahale A, Ghosh B, Kulkarni OP. Pharmacological blockade of HDAC3 accelerates diabetic wound healing by regulating macrophage activation. Life Sci 2023; 321:121574. [PMID: 36931496 DOI: 10.1016/j.lfs.2023.121574] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
AIMS Here, we report the effect of histone deacetylase 3 (HDAC3) inhibition associated with macrophage activation, IL-1β expression, angiogenesis and wound healing in diabetic mice. MAIN METHODS To determine the expression of HDAC3 in diabetic mice wounds, hyperglycemia was induced in C57BL/6 mice with streptozotocin followed by induction of 6 mm wounds. To understand the effect of HDAC3 selective inhibitor, BG45, wound tissues were isolated for analysing M1/M2 markers expression, immune cells infiltration, angiogenesis and healing factors expression. CD11b+F4/80+ cells were sorted from the wound tissues and analysed for the expression of M1/M2 markers using RT-qPCR and flow cytometer. In cell based assays, HDAC3 expression was measured in macrophages stimulated with high glucose (HG) plus LPS. Macrophages treated with BG45 and HG + LPS were analysed for the expression of pro-IL-1β, mature IL-1β, oxidative stress and pro-inflammatory (M1) and anti-inflammatory (M2) factors. KEY FINDINGS HDAC3 was found to be upregulated in impaired diabetic mice wounds and in macrophages stimulated with HG + LPS. Topical application of BG45 loaded gel accelerated the wound healing in diabetic mice and was evident by improved expression of Collagen-1A, IL-10, TGF-β, and angiogenesis (CD31, VEGF). BG45 treatment decreased the expression of IL-1β, TNF-α, and IL-6 (M1 phenotype), reduced oxidative stress and promoted the expression of Arginase-1 and YM1/2 (M2 phenotype) in macrophages treated with HG + LPS. BG45 also improved the expression of CD11b+F4/80+CD206+ cells in wound tissues and reduced expression of inflammatory markers. SIGNIFICANCE HDAC3 is upregulated in diabetic mice wounds and HDAC3 selective inhibitor promotes the wound healing by regulating macrophage activation, angiogenesis and IL-1β.
Collapse
Affiliation(s)
- Kalyani Karnam
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Kavitha Sedmaki
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Pravesh Sharma
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Ashutosh Mahale
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Balaram Ghosh
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India; Epigenetic Research Laboratory, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India
| | - Onkar Prakash Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, India.
| |
Collapse
|
13
|
Jogdeo CM, Panja S, Kanvinde S, Kapoor E, Siddhanta K, Oupický D. Advances in Lipid-Based Codelivery Systems for Cancer and Inflammatory Diseases. Adv Healthc Mater 2023; 12:e2202400. [PMID: 36453542 PMCID: PMC10023350 DOI: 10.1002/adhm.202202400] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/13/2022] [Indexed: 12/03/2022]
Abstract
Combination therapy targeting multiple therapeutic targets is a favorable strategy to achieve better therapeutic outcomes in cancer and inflammatory diseases. Codelivery is a subfield of drug delivery that aims to achieve combined delivery of diverse therapeutic cargoes within the same delivery system, thereby ensuring delivery to the same site and providing an opportunity to tailor the release kinetics as desired. Among the wide range of materials being investigated in the design of codelivery systems, lipids have stood out on account of their low toxicity, biocompatibility, and ease of formulation scale-up. This review highlights the advances of the last decade in lipid-based codelivery systems focusing on the codelivery of drug-drug, drug-nucleic acid, nucleic acid-nucleic acid, and protein therapeutic-based combinations for targeted therapy in cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Chinmay M. Jogdeo
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Sudipta Panja
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Shrey Kanvinde
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Ekta Kapoor
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Kasturi Siddhanta
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - David Oupický
- Center for Drug Delivery and NanomedicineDepartment of Pharmaceutical SciencesUniversity of Nebraska Medical CenterOmahaNE68198USA
| |
Collapse
|
14
|
Fluro-Protein C-Phycocyanin Docked Silver Nanocomposite Accelerates Cell Migration through NFĸB Signaling Pathway. Int J Mol Sci 2023; 24:ijms24043184. [PMID: 36834597 PMCID: PMC9962756 DOI: 10.3390/ijms24043184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/23/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Currently, there is a great demand for the development of nanomedicine aided wound tissue regeneration via silver doped nanoceuticals. Unfortunately, very little research is being carried out on antioxidants-doped silver nanometals and their interaction on the signaling axis during the bio-interface mechanism. In this study, c-phycocyanin primed silver nano hybrids (AgcPCNP) were prepared and analyzed for properties such as cytotoxicity, metal decay, nanoconjugate stability, size expansion, and antioxidant features. Fluctuations in the expression of marker genes during cell migration phenomena in in vitro wound healing scenarios were also validated. Studies revealed that physiologically relevant ionic solutions did not exhibit any adverse effects on the nanoconjugate stability. However, acidic, alkali, and ethanol solutions completely denatured the AgcPCNP conjugates. Signal transduction RT2PCR array demonstrated that genes associated with NFĸB- and PI3K-pathways were significantly (p < 0.5%) altered between AgcPCNP and AgNP groups. Specific inhibitors of NFĸB (Nfi) and PI3K (LY294002) pathways confirmed the involvement of NFĸB signaling axes. In vitro wound healing assay demonstrated that NFĸB pathway plays a prime role in the fibroblast cell migration. In conclusion, the present investigation revealed that surface functionalized AgcPCNP accelerated the fibroblast cell migration and can be further explored for wound healing biomedical applications.
Collapse
|
15
|
Multiple Genetic Loci Associated with Pug Dog Thoracolumbar Myelopathy. Genes (Basel) 2023; 14:genes14020385. [PMID: 36833311 PMCID: PMC9957375 DOI: 10.3390/genes14020385] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Pug dogs with thoracolumbar myelopathy (PDM) present with a specific clinical phenotype that includes progressive pelvic limb ataxia and paresis, commonly accompanied by incontinence. Vertebral column malformations and lesions, excessive scar tissue of the meninges, and central nervous system inflammation have been described. PDM has a late onset and affects more male than female dogs. The breed-specific presentation of the disorder suggests that genetic risk factors are involved in the disease development. To perform a genome-wide search for PDM-associated loci, we applied a Bayesian model adapted for mapping complex traits (BayesR) and a cross-population extended haplotype homozygosity test (XP-EHH) in 51 affected and 38 control pugs. Nineteen associated loci (harboring 67 genes in total, including 34 potential candidate genes) and three candidate regions under selection (with four genes within or next to the signal) were identified. The multiple candidate genes identified have implicated functions in bone homeostasis, fibrotic scar tissue, inflammatory responses, or the formation, regulation, and differentiation of cartilage, suggesting the potential relevance of these processes to the pathogenesis of PDM.
Collapse
|
16
|
Longo V, Aloi N, Lo Presti E, Fiannaca A, Longo A, Adamo G, Urso A, Meraviglia S, Bongiovanni A, Cibella F, Colombo P. Impact of the flame retardant 2,2'4,4'-tetrabromodiphenyl ether (PBDE-47) in THP-1 macrophage-like cell function via small extracellular vesicles. Front Immunol 2023; 13:1069207. [PMID: 36685495 PMCID: PMC9852912 DOI: 10.3389/fimmu.2022.1069207] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/13/2022] [Indexed: 01/07/2023] Open
Abstract
2,2'4,4'-tetrabromodiphenyl ether (PBDE-47) is one of the most widespread environmental brominated flame-retardant congeners which has also been detected in animal and human tissues. Several studies have reported the effects of PBDEs on different health issues, including neurobehavioral and developmental disorders, reproductive health, and alterations of thyroid function. Much less is known about its immunotoxicity. The aim of our study was to investigate the effects that treatment of THP-1 macrophage-like cells with PBDE-47 could have on the content of small extracellular vesicles' (sEVs) microRNA (miRNA) cargo and their downstream effects on bystander macrophages. To achieve this, we purified sEVs from PBDE-47 treated M(LPS) THP-1 macrophage-like cells (sEVsPBDE+LPS) by means of ultra-centrifugation and characterized their miRNA cargo by microarray analysis detecting the modulation of 18 miRNAs. Furthermore, resting THP-1 derived M(0) macrophage-like cells were cultured with sEVsPBDE+LPS, showing that the treatment reshaped the miRNA profiles of 12 intracellular miRNAs. This dataset was studied in silico, identifying the biological pathways affected by these target genes. This analysis identified 12 pathways all involved in the maturation and polarization of macrophages. Therefore, to evaluate whether sEVsPBDE+LPS can have some immunomodulatory activity, naïve M(0) THP-1 macrophage-like cells cultured with purified sEVsPBDE+LPS were studied for IL-6, TNF-α and TGF-β mRNAs expression and immune stained with the HLA-DR, CD80, CCR7, CD38 and CD209 antigens and analyzed by flow cytometry. This analysis showed that the PBDE-47 treatment does not induce the expression of specific M1 and M2 cytokine markers of differentiation and may have impaired the ability to make immunological synapses and present antigens, down-regulating the expression of HLA-DR and CD209 antigens. Overall, our study supports the model that perturbation of miRNA cargo by PBDE-47 treatment contributes to the rewiring of cellular regulatory pathways capable of inducing perturbation of differentiation markers on naïve resting M(0) THP-1 macrophage-like cells.
Collapse
Affiliation(s)
- Valeria Longo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Noemi Aloi
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Elena Lo Presti
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Antonino Fiannaca
- High Performance Computing and Networking Institute, National Research Council of Italy (ICAR-CNR), Palermo, Italy
| | - Alessandra Longo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Giorgia Adamo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Alfonso Urso
- High Performance Computing and Networking Institute, National Research Council of Italy (ICAR-CNR), Palermo, Italy
| | - Serena Meraviglia
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Antonella Bongiovanni
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Fabio Cibella
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy
| | - Paolo Colombo
- Institute for Biomedical Research and Innovation, National Research Council of Italy (IRIB-CNR), Palermo, Italy,*Correspondence: Paolo Colombo,
| |
Collapse
|
17
|
Bae S, Kim K, Kang K, Kim H, Lee M, Oh B, Kaneko K, Ma S, Choi JH, Kwak H, Lee EY, Park SH, Park-Min KH. RANKL-responsive epigenetic mechanism reprograms macrophages into bone-resorbing osteoclasts. Cell Mol Immunol 2023; 20:94-109. [PMID: 36513810 PMCID: PMC9794822 DOI: 10.1038/s41423-022-00959-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/03/2022] [Indexed: 12/15/2022] Open
Abstract
Monocyte/macrophage lineage cells are highly plastic and can differentiate into various cells under different environmental stimuli. Bone-resorbing osteoclasts are derived from the monocyte/macrophage lineage in response to receptor activator of NF-κB ligand (RANKL). However, the epigenetic signature contributing to the fate commitment of monocyte/macrophage lineage differentiation into human osteoclasts is largely unknown. In this study, we identified RANKL-responsive human osteoclast-specific superenhancers (SEs) and SE-associated enhancer RNAs (SE-eRNAs) by integrating data obtained from ChIP-seq, ATAC-seq, nuclear RNA-seq and PRO-seq analyses. RANKL induced the formation of 200 SEs, which are large clusters of enhancers, while suppressing 148 SEs in macrophages. RANKL-responsive SEs were strongly correlated with genes in the osteoclastogenic program and were selectively increased in human osteoclasts but marginally presented in osteoblasts, CD4+ T cells, and CD34+ cells. In addition to the major transcription factors identified in osteoclasts, we found that BATF binding motifs were highly enriched in RANKL-responsive SEs. The depletion of BATF1/3 inhibited RANKL-induced osteoclast differentiation. Furthermore, we found increased chromatin accessibility in SE regions, where RNA polymerase II was significantly recruited to induce the extragenic transcription of SE-eRNAs, in human osteoclasts. Knocking down SE-eRNAs in the vicinity of the NFATc1 gene diminished the expression of NFATc1, a major regulator of osteoclasts, and osteoclast differentiation. Inhibiting BET proteins suppressed the formation of some RANKL-responsive SEs and NFATc1-associated SEs, and the expression of SE-eRNA:NFATc1. Moreover, SE-eRNA:NFATc1 was highly expressed in the synovial macrophages of rheumatoid arthritis patients exhibiting high-osteoclastogenic potential. Our genome-wide analysis revealed RANKL-inducible SEs and SE-eRNAs as osteoclast-specific signatures, which may contribute to the development of osteoclast-specific therapeutic interventions.
Collapse
Affiliation(s)
- Seyeon Bae
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Kibyeong Kim
- Department of Biological Science, Ulsan National Institute of Science & Technology (UNIST), Ulsan, 44919, Republic of Korea
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Keunsoo Kang
- Department of Microbiology, Dankook University, Cheonan, 3116, Republic of Korea
| | - Haemin Kim
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Minjoon Lee
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Brian Oh
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Kaichi Kaneko
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA
| | - Sungkook Ma
- Department of Biological Science, Ulsan National Institute of Science & Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jae Hoon Choi
- Department of Life Science, College of Natural Sciences, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Hojoong Kwak
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, USA
| | - Eun Young Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea.
| | - Sung Ho Park
- Department of Biological Science, Ulsan National Institute of Science & Technology (UNIST), Ulsan, 44919, Republic of Korea.
| | - Kyung-Hyun Park-Min
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, 10021, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
- BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, 10021, USA.
| |
Collapse
|
18
|
Lin D, Xu W, Hong P, Wu C, Zhang Z, Zhang S, Xing L, Yang B, Zhou W, Xiao Q, Wang J, Wang C, He Y, Chen X, Cao X, Man J, Reheman A, Wu X, Hao X, Hu Z, Chen C, Cao Z, Yin R, Fu ZF, Zhou R, Teng Z, Li G, Cao G. Decoding the spatial chromatin organization and dynamic epigenetic landscapes of macrophage cells during differentiation and immune activation. Nat Commun 2022; 13:5857. [PMID: 36195603 PMCID: PMC9532393 DOI: 10.1038/s41467-022-33558-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 09/22/2022] [Indexed: 11/09/2022] Open
Abstract
Immunocytes dynamically reprogram their gene expression profiles during differentiation and immunoresponse. However, the underlying mechanism remains elusive. Here, we develop a single-cell Hi-C method and systematically delineate the 3D genome and dynamic epigenetic atlas of macrophages during these processes. We propose "degree of disorder" to measure genome organizational patterns inside topologically-associated domains, which is correlated with the chromatin epigenetic states, gene expression, and chromatin structure variability in individual cells. Furthermore, we identify that NF-κB initiates systematic chromatin conformation reorganization upon Mycobacterium tuberculosis infection. The integrated Hi-C, eQTL, and GWAS analysis depicts the atlas of the long-range target genes of mycobacterial disease susceptible loci. Among these, the SNP rs1873613 is located in the anchor of a dynamic chromatin loop with LRRK2, whose inhibitor AdoCbl could be an anti-tuberculosis drug candidate. Our study provides comprehensive resources for the 3D genome structure of immunocytes and sheds insights into the order of genome organization and the coordinated gene transcription during immunoresponse.
Collapse
Affiliation(s)
- Da Lin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Bio-Medicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Weize Xu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ping Hong
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, China
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, 3D Genomics Research Center, Huazhong Agricultural University, Wuhan, China
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Chengchao Wu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhihui Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Siheng Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lingyu Xing
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Bing Yang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wei Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qin Xiao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Bio-Medicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Jinyue Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Bio-Medicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Cong Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yu He
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xi Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaojian Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jiangwei Man
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Aikebaier Reheman
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Animal Science and Technology, Tarim University, Alar, China
| | - Xiaofeng Wu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xingjie Hao
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe Hu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Chunli Chen
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region, Guizhou University, Guiyang, China
| | - Zimeng Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- College of Bio-Medicine and Health, Huazhong Agricultural University, Wuhan, China
- College of Animal Sciences, Yangtze River University, Jingzhou, China
| | - Rong Yin
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhen F Fu
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Rong Zhou
- Dapartment of Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhaowei Teng
- The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Guoliang Li
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan, China.
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, 3D Genomics Research Center, Huazhong Agricultural University, Wuhan, China.
- College of Informatics, Huazhong Agricultural University, Wuhan, China.
| | - Gang Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
- College of Bio-Medicine and Health, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
19
|
Tuerxun K, Midtbö K, Särndahl E, Vorontsov E, Karlsson R, Persson A, Kruse R, Eklund D, the X‐HiDE Consortium. Cytokine responses to LPS in reprogrammed monocytes are associated with the transcription factor PU.1. J Leukoc Biol 2022; 112:679-692. [PMID: 35285058 PMCID: PMC9790682 DOI: 10.1002/jlb.3a0421-216r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 01/21/2022] [Accepted: 01/30/2022] [Indexed: 12/30/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are functionally immunosuppressive cells that arise and expand during extensive inflammatory conditions by increased hematopoietic output or reprogramming of immune cells. In sepsis, an increase of circulating MDSCs is associated with adverse outcomes, but unique traits that can be used to identify increased activity of MDSCs are lacking. By using endotoxin tolerance as a model of sepsis-induced monocytic MDSCs (M-MDSC-like cells), this study aims to identify the mediator and transcriptional regulator profile associated with M-MDSC activity. After analyzing 180 inflammation-associated proteins, a profile of differentially expressed cytokines was found in M-MDSC-like cells versus normal monocytes stimulated with LPS. These cytokines were associated with 5 candidate transcription factors, where particularly PU.1 showed differential expression on both transcriptional and protein levels in M-MDSC-like cells. Furthermore, inhibition of PU.1 led to increased production of CXCL5 and CCL8 in M-MDSC-like cells indicating its role in regulating the ability of M-MDSC-like cells to recruit other immune cells. Taken together, the study identifies a unique profile in the pattern of immune mediators defining M-MDSC activity upon LPS stimulation, which offers a functional link to their contribution to immunosuppression.
Collapse
Affiliation(s)
- Kedeye Tuerxun
- Faculty of Medicine and Health, School of Medical SciencesÖrebro UniversityÖrebroSweden,Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and HealthÖrebro UniversityÖrebroSweden
| | - Kristine Midtbö
- Faculty of Medicine and Health, School of Medical SciencesÖrebro UniversityÖrebroSweden,Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and HealthÖrebro UniversityÖrebroSweden
| | - Eva Särndahl
- Faculty of Medicine and Health, School of Medical SciencesÖrebro UniversityÖrebroSweden,Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and HealthÖrebro UniversityÖrebroSweden
| | - Egor Vorontsov
- Proteomics Core Facility, Sahlgrenska AcademyUniversity of GothenburgSweden
| | - Roger Karlsson
- Department of Infectious Diseases, Institute of BiomedicineSahlgrenska Academy of the University of GothenburgSweden,Department of Clinical MicrobiologySahlgrenska University Hospital, Region Västra GötalandSweden,Nanoxis Consulting ABGothenburgSweden
| | - Alexander Persson
- Faculty of Medicine and Health, School of Medical SciencesÖrebro UniversityÖrebroSweden,Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and HealthÖrebro UniversityÖrebroSweden
| | - Robert Kruse
- Faculty of Medicine and Health, School of Medical SciencesÖrebro UniversityÖrebroSweden,Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and HealthÖrebro UniversityÖrebroSweden,Department of Clinical Research Laboratory, Faculty of Medicine and HealthÖrebro UniversityÖrebroSweden
| | - Daniel Eklund
- Faculty of Medicine and Health, School of Medical SciencesÖrebro UniversityÖrebroSweden,Inflammatory Response and Infection Susceptibility Centre (iRiSC), Faculty of Medicine and HealthÖrebro UniversityÖrebroSweden
| | | |
Collapse
|
20
|
Wang Y, Liu X, Xia P, Li Z, FuChen X, Shen Y, Yu P, Zhang J. The Regulatory Role of MicroRNAs on Phagocytes: A Potential Therapeutic Target for Chronic Diseases. Front Immunol 2022; 13:901166. [PMID: 35634335 PMCID: PMC9130478 DOI: 10.3389/fimmu.2022.901166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/19/2022] [Indexed: 11/27/2022] Open
Abstract
An effective acute inflammatory response results in the elimination of infectious microorganisms, followed by a smooth transition to resolution and repair. During the inflammatory response, neutrophils play a crucial role in antimicrobial defense as the first cells to reach the site of infection damage. However, if the neutrophils that have performed the bactericidal effect are not removed in time, the inflammatory response will not be able to subside. Anti-inflammatory macrophages are the main scavengers of neutrophils and can promote inflammation towards resolution. MicroRNAs (miRNAs) have great potential as clinical targeted therapy and have attracted much attention in recent years. This paper summarizes the involvement of miRNAs in the process of chronic diseases such as atherosclerosis, rheumatoid arthritis and systemic lupus erythematosus by regulating lipid metabolism, cytokine secretion, inflammatory factor synthesis and tissue repair in two types of cells. This will provide a certain reference for miRNA-targeted treatment of chronic diseases.
Collapse
Affiliation(s)
- Yongbo Wang
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Xingyu Liu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Panpan Xia
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Zhangwang Li
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Xinxi FuChen
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Yunfeng Shen
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Jiangxi, China
| |
Collapse
|
21
|
Fraschilla I, Amatullah H, Jeffrey KL. One genome, many cell states: epigenetic control of innate immunity. Curr Opin Immunol 2022; 75:102173. [PMID: 35405493 PMCID: PMC9081230 DOI: 10.1016/j.coi.2022.102173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/23/2022] [Accepted: 02/23/2022] [Indexed: 12/15/2022]
Abstract
A hallmark of the innate immune system is its ability to rapidly initiate short-lived or sustained transcriptional programs in a cell-specific and pathogen-specific manner that is dependent on dynamic chromatin states. Much of the epigenetic landscape is set during cellular differentiation; however, pathogens and other environmental cues also induce changes in chromatin that can either promote tolerance or 'train' innate immune cells for amplified secondary responses. We review chromatin processes that enable innate immune cell differentiation and functional transcriptional responses in naive or experienced cells, in concert with signal transduction and cellular metabolic shifts. We discuss how immune chromatin mechanisms are maladapted in disease and novel therapeutic approaches for cellular reprogramming.
Collapse
Affiliation(s)
- Isabella Fraschilla
- Division of Gastroenterology and Center for the Study of Inflammatory Bowel Disease, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA; Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Hajera Amatullah
- Division of Gastroenterology and Center for the Study of Inflammatory Bowel Disease, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Kate L Jeffrey
- Division of Gastroenterology and Center for the Study of Inflammatory Bowel Disease, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA; Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
22
|
Miller GE, Chen E, Finegood E, Shimbo D, Cole SW. Prospective associations between neighborhood violence and monocyte pro-inflammatory transcriptional activity in children. Brain Behav Immun 2022; 100:1-7. [PMID: 34800620 PMCID: PMC8766930 DOI: 10.1016/j.bbi.2021.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/18/2021] [Accepted: 11/05/2021] [Indexed: 02/03/2023] Open
Abstract
Individuals exposed to persistent neighborhood violence are at increased risk for developing mental and physical health problems across the lifespan. The biological mechanisms underlying this phenomenon are not well understood. Thus, we examined the relationship between children's exposure to neighborhood violence and inflammatory activity, a process involved in the pathogenesis of multiple health problems. 236 children from the Chicago area participated in a two-year longitudinal study (mean age at baseline, 13.9 years; 67% female; 39% White, 34% Black, 33% Hispanic). Neighborhood violence was measured as the homicide frequency in a child's Census block group in the five years before study entry. Fasting blood was drawn at study entry and two years later (in eighth and tenth grade). The blood was used to quantify protein biomarkers of systemic inflammatory activity and perform genome-wide expression profiling of isolated monocytes. Neighborhood violence was associated with higher systemic inflammatory activity at both assessments. It also was associated with a monocyte transcriptional profile indicative of increased signaling along the nuclear factor-kappa B (NF-κB) and activator protein 1 (AP-1) control pathways, which are key orchestrators of pro-inflammatory effector functions. Neighborhood violence also was associated with transcriptional indications of higher beta-adrenergic and lower glucocorticoid signaling, which could function as neuroendocrine conduits linking threatening experiences with inflammatory activity. Neighborhood violence was not associated with two-year changes in protein biomarkers, although it did presage a transcriptional profile indicative of increasing AP-1 and declining glucocorticoid signaling over follow-up. Collectively, these observations highlight cellular and molecular pathways that could underlie health risks associated with neighborhood violence.
Collapse
Affiliation(s)
- Gregory E Miller
- Institute for Policy Research and Department of Psychology, Northwestern University, Evanston, IL, USA.
| | - Edith Chen
- Institute for Policy Research and Department of Psychology, Northwestern University, Evanston, IL, USA
| | - Eric Finegood
- Institute for Policy Research and Department of Psychology, Northwestern University, Evanston, IL, USA
| | - Daichi Shimbo
- Columbia Hypertension Center and Lab, Department of Medicine, Columbia University, New York, NY, USA
| | - Steve W Cole
- Cousins Center for Psychoneuroimmunology and Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
23
|
Fang JS, Hultgren NW, Hughes CCW. Regulation of Partial and Reversible Endothelial-to-Mesenchymal Transition in Angiogenesis. Front Cell Dev Biol 2021; 9:702021. [PMID: 34692672 PMCID: PMC8529039 DOI: 10.3389/fcell.2021.702021] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022] Open
Abstract
During development and in several diseases, endothelial cells (EC) can undergo complete endothelial-to-mesenchymal transition (EndoMT or EndMT) to generate endothelial-derived mesenchymal cells. Emerging evidence suggests that ECs can also undergo a partial EndoMT to generate cells with intermediate endothelial- and mesenchymal-character. This partial EndoMT event is transient, reversible, and supports both developmental and pathological angiogenesis. Here, we discuss possible regulatory mechanisms that may control the EndoMT program to dictate whether cells undergo complete or partial mesenchymal transition, and we further consider how these pathways might be targeted therapeutically in cancer.
Collapse
Affiliation(s)
- Jennifer S. Fang
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Nan W. Hultgren
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Christopher C. W. Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
24
|
George TB, Strawn NK, Leviyang S. Tree-Based Co-Clustering Identifies Chromatin Accessibility Patterns Associated With Hematopoietic Lineage Structure. Front Genet 2021; 12:707117. [PMID: 34659332 PMCID: PMC8517275 DOI: 10.3389/fgene.2021.707117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 09/14/2021] [Indexed: 01/21/2023] Open
Abstract
Chromatin accessibility, as measured by ATACseq, varies between hematopoietic cell types in different lineages of the hematopoietic differentiation tree, e.g. T cells vs. B cells, but methods that associate variation in chromatin accessibility to the lineage structure of the differentiation tree are lacking. Using an ATACseq dataset recently published by the ImmGen consortium, we construct associations between chromatin accessibility and hematopoietic cell types using a novel co-clustering approach that accounts for the structure of the hematopoietic, differentiation tree. Under a model in which all loci and cell types within a co-cluster have a shared accessibility state, we show that roughly 80% of cell type associated accessibility variation can be captured through 12 cell type clusters and 20 genomic locus clusters, with the cell type clusters reflecting coherent components of the differentiation tree. Using publicly available ChIPseq datasets, we show that our clustering reflects transcription factor binding patterns with implications for regulation across cell types. We show that traditional methods such as hierarchical and kmeans clusterings lead to cell type clusters that are more dispersed on the tree than our tree-based algorithm. We provide a python package, chromcocluster, that implements the algorithms presented.
Collapse
Affiliation(s)
| | | | - Sivan Leviyang
- Department of Mathematics and Statistics, Georgetown University, Washington, DC, United States
| |
Collapse
|
25
|
Leviyang S. Interferon stimulated binding of ISRE is cell type specific and is predicted by homeostatic chromatin state. Cytokine X 2021; 3:100056. [PMID: 34409284 PMCID: PMC8361084 DOI: 10.1016/j.cytox.2021.100056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
IFN stimulated binding of ISRE by ISGF3 is cell specific, particularly for ISRE in enhancer regions. IFN stimulated binding of ISRE in enhancer regions associates with differential expression. The homeostatic, chromatin state of an ISRE is predictive of IFN stimulated binding.
The type I interferon (IFN) signaling pathway involves binding of the transcription factor ISGF3 to IFN-stimulated response elements, ISREs. Gene expression under IFN stimulation is known to vary across cell types, but variation in ISGF3 binding to ISRE across cell types has not been characterized. We examined ISRE binding patterns under IFN stimulation across six cell types using existing ChIPseq datasets. We find that ISRE binding is largely cell specific for ISREs distal to transcription start sites (TSS) and largely conserved across cell types for ISREs proximal to TSS. We show that bound ISRE distal to TSS associate with differential expression of ISGs, although at weaker levels than bound ISRE proximal to TSS. Using existing ATACseq and ChIPseq datasets, we show that the chromatin state of ISRE at homeostasis is cell type specific and is predictive of cell specific, ISRE binding under IFN stimulation. Our results support a model in which the chromatin state of ISRE in enhancer elements is modulated in a cell type specific manner at homeostasis, leading to cell type specific differences in ISRE binding patterns under IFN stimulation.
Collapse
|
26
|
Piatek P, Tarkowski M, Namiecinska M, Riva A, Wieczorek M, Michlewska S, Dulska J, Domowicz M, Kulińska-Michalska M, Lewkowicz N, Lewkowicz P. H3K4me3 Histone ChIP-Seq Analysis Reveals Molecular Mechanisms Responsible for Neutrophil Dysfunction in HIV-Infected Individuals. Front Immunol 2021; 12:682094. [PMID: 34335583 PMCID: PMC8320512 DOI: 10.3389/fimmu.2021.682094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/25/2021] [Indexed: 01/15/2023] Open
Abstract
Peripheral neutrophils in HIV-infected individuals are characterized by impairment of chemotaxis, phagocytosis, bactericidal activity, and oxidative burst ability regardless of whether patients are receiving antiretroviral therapy or not. Neutrophil dysfunction leads not only to increased susceptibility to opportunistic infections but also to tissue damage through the release of reactive oxygen species (ROS), proteases, and other potentially harmful effector molecules contributing to AIDS progression. In this study, we demonstrated high levels of histone H3 lysine K4 trimethylated (H3K4me3) and dysregulation of DNA transcription in circulating neutrophils of HIV-infected subjects. This dysregulation was accompanied by a deficient response of neutrophils to LPS, impaired cytokine/chemokine/growth factor synthesis, and increased apoptosis. Chromatin immunoprecipitation sequencing (ChIPseq) H3K4me3 histone analysis revealed that the most spectacular abnormalities were observed in the exons, introns, and promoter-TSS regions. Bioinformatic analysis of Gene Ontology, including biological processes, molecular function, and cellular components, demonstrated that the main changes were related to the genes responsible for cell activation, cytokine production, adhesive molecule expression, histone remodeling via upregulation of methyltransferase process, and downregulation of NF-κB transcription factor in canonical pathways. Abnormalities within H3K4me3 implicated LPS-mediated NF-κB canonical activation pathway that was a result of low amounts of κB DNA sites within histone H3K4me3, low NF-κB (p65 RelA) and TLR4 mRNA expression, and reduced free NF-κB (p65 RelA) accumulation in the nucleus. Genome-wide survey of H3K4me3 provided evidence that chromatin modifications lead to an impairment within the canonical NF-κB cell activation pathway causing the neutrophil dysfunction observed in HIV-infected individuals.
Collapse
Affiliation(s)
- Paweł Piatek
- Department of Neurology, Laboratory of Neuroimmunology, Medical University of Lodz, Lodz, Poland
| | - Maciej Tarkowski
- Department of Biomedical and Clinical Sciences, 'Luigi Sacco', University of Milan, Milan, Italy
| | - Magdalena Namiecinska
- Department of Neurology, Laboratory of Neuroimmunology, Medical University of Lodz, Lodz, Poland
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences, 'Luigi Sacco', University of Milan, Milan, Italy
| | - Marek Wieczorek
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Sylwia Michlewska
- Laboratory of Microscopic Imaging and Specialized Biological Techniques, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | | | - Małgorzata Domowicz
- Department of Neurology, Laboratory of Neuroimmunology, Medical University of Lodz, Lodz, Poland
| | | | - Natalia Lewkowicz
- Department of Periodontology and Oral Mucosal Diseases, Medical University of Lodz, Lodz, Poland
| | - Przemysław Lewkowicz
- Department of Neurology, Laboratory of Neuroimmunology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
27
|
Onodera A, González-Avalos E, Lio CWJ, Georges RO, Bellacosa A, Nakayama T, Rao A. Roles of TET and TDG in DNA demethylation in proliferating and non-proliferating immune cells. Genome Biol 2021; 22:186. [PMID: 34158086 PMCID: PMC8218415 DOI: 10.1186/s13059-021-02384-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/21/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND TET enzymes mediate DNA demethylation by oxidizing 5-methylcytosine (5mC) in DNA to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC). Since these oxidized methylcytosines (oxi-mCs) are not recognized by the maintenance methyltransferase DNMT1, DNA demethylation can occur through "passive," replication-dependent dilution when cells divide. A distinct, replication-independent ("active") mechanism of DNA demethylation involves excision of 5fC and 5caC by the DNA repair enzyme thymine DNA glycosylase (TDG), followed by base excision repair. RESULTS Here by analyzing inducible gene-disrupted mice, we show that DNA demethylation during primary T cell differentiation occurs mainly through passive replication-dependent dilution of all three oxi-mCs, with only a negligible contribution from TDG. In addition, by pyridine borane sequencing (PB-seq), a simple recently developed method that directly maps 5fC/5caC at single-base resolution, we detect the accumulation of 5fC/5caC in TDG-deleted T cells. We also quantify the occurrence of concordant demethylation within and near enhancer regions in the Il4 locus. In an independent system that does not involve cell division, macrophages treated with liposaccharide accumulate 5hmC at enhancers and show altered gene expression without DNA demethylation; loss of TET enzymes disrupts gene expression, but loss of TDG has no effect. We also observe that mice with long-term (1 year) deletion of Tdg are healthy and show normal survival and hematopoiesis. CONCLUSIONS We have quantified the relative contributions of TET and TDG to cell differentiation and DNA demethylation at representative loci in proliferating T cells. We find that TET enzymes regulate T cell differentiation and DNA demethylation primarily through passive dilution of oxi-mCs. In contrast, while we observe a low level of active, replication-independent DNA demethylation mediated by TDG, this process does not appear to be essential for immune cell activation or differentiation.
Collapse
Affiliation(s)
- Atsushi Onodera
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
- Institute for Global Prominent Research, Chiba University, 1-33, Yayoicho, Inage-ku, Chiba, 263-8522, Japan
| | - Edahí González-Avalos
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Chan-Wang Jerry Lio
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
- Present address: Department of Microbial Infection and Immunity, Ohio State University, 460 W 12th Ave, Columbus, OH, 43210, USA
| | - Romain O Georges
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA
| | - Alfonso Bellacosa
- Cancer Signaling and Epigenetics Program & Cancer Epigenetics Institute, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
- AMED-CREST, AMED, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Anjana Rao
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA, 92037, USA.
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA.
| |
Collapse
|
28
|
Rothenberg EV. Logic and lineage impacts on functional transcription factor deployment for T-cell fate commitment. Biophys J 2021; 120:4162-4181. [PMID: 33838137 PMCID: PMC8516641 DOI: 10.1016/j.bpj.2021.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/22/2021] [Accepted: 04/02/2021] [Indexed: 11/19/2022] Open
Abstract
Transcription factors are the major agents that read the regulatory sequence information in the genome to initiate changes in expression of specific genes, both in development and in physiological activation responses. Their actions depend on site-specific DNA binding and are largely guided by their individual DNA target sequence specificities. However, their action is far more conditional in a real developmental context than would be expected for simple reading of local genomic DNA sequence, which is common to all cells in the organism. They are constrained by slow-changing chromatin states and by interactions with other transcription factors, which affect their occupancy patterns of potential sites across the genome. These mechanisms lead to emergent discontinuities in function even for transcription factors with minimally changing expression. This is well revealed by diverse lineages of blood cells developing throughout life from hematopoietic stem cells, which use overlapping combinations of transcription factors to drive strongly divergent gene regulation programs. Here, using development of T lymphocytes from hematopoietic multipotent progenitor cells as a focus, recent evidence is reviewed on how binding specificity and dynamics, transcription factor cooperativity, and chromatin state changes impact the effective regulatory functions of key transcription factors including PU.1, Runx1, Notch-RBPJ, and Bcl11b.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California.
| |
Collapse
|
29
|
Song R, Gao Y, Dozmorov I, Malladi V, Saha I, McDaniel MM, Parameswaran S, Liang C, Arana C, Zhang B, Wakeland B, Zhou J, Weirauch MT, Kottyan LC, Wakeland EK, Pasare C. IRF1 governs the differential interferon-stimulated gene responses in human monocytes and macrophages by regulating chromatin accessibility. Cell Rep 2021; 34:108891. [PMID: 33761354 PMCID: PMC8300000 DOI: 10.1016/j.celrep.2021.108891] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/27/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023] Open
Abstract
Myeloid lineage cells use TLRs to recognize and respond to diverse microbial ligands. Although unique transcription factors dictate the outcome of specific TLR signaling, whether lineage-specific differences exist to further modulate the quality of TLR-induced inflammation remains unclear. Comprehensive analysis of global gene transcription in human monocytes, monocyte-derived macrophages, and monocyte-derived dendritic cells stimulated with various TLR ligands identifies multiple lineage-specific, TLR-responsive gene programs. Monocytes are hyperresponsive to TLR7/8 stimulation that correlates with the higher expression of the receptors. While macrophages and monocytes express similar levels of TLR4, macrophages, but not monocytes, upregulate interferon-stimulated genes (ISGs) in response to TLR4 stimulation. We find that TLR4 signaling in macrophages uniquely engages transcription factor IRF1, which facilitates the opening of ISG loci for transcription. This study provides a critical mechanistic basis for lineage-specific TLR responses and uncovers IRF1 as a master regulator for the ISG transcriptional program in human macrophages.
Collapse
Affiliation(s)
- Ran Song
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yajing Gao
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Igor Dozmorov
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Venkat Malladi
- Bioinformatics Core Facility, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Irene Saha
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Margaret M McDaniel
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sreeja Parameswaran
- Center for Autoimmune Genetics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Chaoying Liang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Carlos Arana
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bo Zhang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Benjamin Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jinchun Zhou
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Matthew T Weirauch
- Center for Autoimmune Genetics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Leah C Kottyan
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Autoimmune Genetics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Edward K Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Chandrashekhar Pasare
- Division of Immunobiology and Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
30
|
Seyedsadjadi N, Grant R. The Potential Benefit of Monitoring Oxidative Stress and Inflammation in the Prevention of Non-Communicable Diseases (NCDs). Antioxidants (Basel) 2020; 10:E15. [PMID: 33375428 PMCID: PMC7824370 DOI: 10.3390/antiox10010015] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
The significant increase in worldwide morbidity and mortality from non-communicable diseases (NCDs) indicates that the efficacy of existing strategies addressing this crisis may need improvement. Early identification of the metabolic irregularities associated with the disease process may be a key to developing early intervention strategies. Unhealthy lifestyle behaviours are well established drivers of the development of several NCDs, but the impact of such behaviours on health can vary considerably between individuals. How can it be determined if an individual's unique set of lifestyle behaviours is producing disease? Accumulating evidence suggests that lifestyle-associated activation of oxidative and inflammatory processes is primary driver of the cell and tissue damage which underpins the development of NCDs. However, the benefit of monitoring subclinical inflammation and oxidative activity has not yet been established. After reviewing relevant studies in this context, we suggest that quantification of oxidative stress and inflammatory biomarkers during the disease-free prodromal stage of NCD development may have clinical relevance as a timely indicator of the presence of subclinical metabolic changes, in the individual, portending the development of disease. Monitoring markers of oxidative and inflammatory activity may therefore enable earlier and more efficient strategies to both prevent NCD development and/or monitor the effectiveness of treatment.
Collapse
Affiliation(s)
- Neda Seyedsadjadi
- Australasian Research Institute, Sydney Adventist Hospital, Sydney, NSW 2076, Australia;
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ross Grant
- Australasian Research Institute, Sydney Adventist Hospital, Sydney, NSW 2076, Australia;
- Department of Pharmacology, School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
- Sydney Adventist Hospital Clinical School, University of Sydney, Sydney, NSW 2076, Australia
| |
Collapse
|
31
|
Ewald AC, Kiernan EA, Roopra AS, Radcliff AB, Timko RR, Baker TL, Watters JJ. Sex- and Region-Specific Differences in the Transcriptomes of Rat Microglia from the Brainstem and Cervical Spinal Cord. J Pharmacol Exp Ther 2020; 375:210-222. [PMID: 32661056 PMCID: PMC7569313 DOI: 10.1124/jpet.120.266171] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
The neural control system underlying breathing is sexually dimorphic with males being more vulnerable to dysfunction. Microglia also display sex differences, and their role in the architecture of brainstem respiratory rhythm circuitry and modulation of cervical spinal cord respiratory plasticity is becoming better appreciated. To further understand the molecular underpinnings of these sex differences, we performed RNA sequencing of immunomagnetically isolated microglia from brainstem and cervical spinal cord of adult male and female rats. We used various bioinformatics tools (Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, Reactome, STRING, MAGICTRICKS) to functionally categorize identified gene sets, as well as to pinpoint common transcriptional gene drivers that may be responsible for the observed transcriptomic differences. We found few sex differences in the microglial transcriptomes derived from the brainstem, but several hundred genes were differentially expressed by sex in cervical spinal microglia. Comparing brainstem and spinal microglia within and between sexes, we found that the major factor guiding transcriptomic differences was central nervous system (CNS) location rather than sex. We further identified key transcriptional drivers that may be responsible for the transcriptomic differences observed between sexes and CNS regions; enhancer of zeste homolog 2 emerged as the predominant driver of the differentially downregulated genes. We suggest that functional gene alterations identified in metabolism, transcription, and intercellular communication underlie critical microglial heterogeneity and sex differences in CNS regions that contribute to respiratory disorders categorized by dysfunction in neural control. These data will also serve as an important resource data base to advance our understanding of innate immune cell contributions to sex differences and the field of respiratory neural control. SIGNIFICANCE STATEMENT: The contributions of central nervous system (CNS) innate immune cells to sexually dimorphic differences in the neural circuitry controlling breathing are poorly understood. We identify key transcriptomic differences, and their transcriptional drivers, in microglia derived from the brainstem and the C3-C6 cervical spinal cord of healthy adult male and female rats. Gene alterations identified in metabolism, gene transcription, and intercellular communication likely underlie critical microglial heterogeneity and sex differences in these key CNS regions that contribute to the neural control of breathing.
Collapse
Affiliation(s)
- Andrea C Ewald
- Departments of Comparative Biosciences (A.C.E., E.A.K., A.B.R., R.R.T., T.L.B., J.J.W.) and Neuroscience (A.S.R.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Elizabeth A Kiernan
- Departments of Comparative Biosciences (A.C.E., E.A.K., A.B.R., R.R.T., T.L.B., J.J.W.) and Neuroscience (A.S.R.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Avtar S Roopra
- Departments of Comparative Biosciences (A.C.E., E.A.K., A.B.R., R.R.T., T.L.B., J.J.W.) and Neuroscience (A.S.R.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Abigail B Radcliff
- Departments of Comparative Biosciences (A.C.E., E.A.K., A.B.R., R.R.T., T.L.B., J.J.W.) and Neuroscience (A.S.R.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Rebecca R Timko
- Departments of Comparative Biosciences (A.C.E., E.A.K., A.B.R., R.R.T., T.L.B., J.J.W.) and Neuroscience (A.S.R.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Tracy L Baker
- Departments of Comparative Biosciences (A.C.E., E.A.K., A.B.R., R.R.T., T.L.B., J.J.W.) and Neuroscience (A.S.R.), University of Wisconsin-Madison, Madison, Wisconsin
| | - Jyoti J Watters
- Departments of Comparative Biosciences (A.C.E., E.A.K., A.B.R., R.R.T., T.L.B., J.J.W.) and Neuroscience (A.S.R.), University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
32
|
Cuevas AG, Ong AD, Carvalho K, Ho T, Chan SW(C, Allen JD, Chen R, Rodgers J, Biba U, Williams DR. Discrimination and systemic inflammation: A critical review and synthesis. Brain Behav Immun 2020; 89:465-479. [PMID: 32688027 PMCID: PMC8362502 DOI: 10.1016/j.bbi.2020.07.017] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 07/01/2020] [Accepted: 07/13/2020] [Indexed: 12/21/2022] Open
Abstract
Exposure to discrimination or unfair treatment has emerged as an important risk factor for illness and disease that disproportionately affects racial and ethnic minorities. Discriminatory experiences may operate like other stressors in that they activate physiological responses that adversely affect the maintenance of homeostasis. Research suggests that inflammation plays a critical role in the pathophysiology of stress-related diseases. Recent findings on discrimination and inflammation are discussed. We highlight limitations in the current evidence and provide recommendations for future studies that seek to examine the association between discrimination and inflammation.
Collapse
Affiliation(s)
- Adolfo G. Cuevas
- Department of Community Health, Tufts University, United States,Corresponding author at: Tufts University, Department of Community Health, 574 Boston Ave, Suite 208, Medford, MA 02155, United States. (A.G. Cuevas)
| | - Anthony D. Ong
- Department of Human Development, Cornell University, United States,Department of Medicine, Weill Cornell Medical College, United States
| | - Keri Carvalho
- Department of Community Health, Tufts University, United States
| | - Thao Ho
- Department of Community Health, Tufts University, United States
| | | | | | - Ruijia Chen
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, United States
| | - Justin Rodgers
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, United States
| | - Ursula Biba
- Department of Community Health, Tufts University, United States
| | - David R. Williams
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, United States,Department of African and African American Studies, Harvard University, United States
| |
Collapse
|
33
|
Tang MS, Miraldi ER, Girgis NM, Bonneau RA, Loke P. Alternative Activation of Macrophages Is Accompanied by Chromatin Remodeling Associated with Lineage-Dependent DNA Shape Features Flanking PU.1 Motifs. THE JOURNAL OF IMMUNOLOGY 2020; 205:1070-1083. [PMID: 32661179 DOI: 10.4049/jimmunol.2000258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/17/2020] [Indexed: 01/04/2023]
Abstract
IL-4 activates macrophages to adopt distinct phenotypes associated with clearance of helminth infections and tissue repair, but the phenotype depends on the cellular lineage of these macrophages. The molecular basis of chromatin remodeling in response to IL-4 stimulation in tissue-resident and monocyte-derived macrophages is not understood. In this study, we find that IL-4 activation of different lineages of peritoneal macrophages in mice is accompanied by lineage-specific chromatin remodeling in regions enriched with binding motifs of the pioneer transcription factor PU.1. PU.1 motif is similarly associated with both tissue-resident and monocyte-derived IL-4-induced accessible regions but has different lineage-specific DNA shape features and predicted cofactors. Mutation studies based on natural genetic variation between C57BL/6 and BALB/c mouse strains indicate that accessibility of these IL-4-induced regions can be regulated through differences in DNA shape without direct disruption of PU.1 motifs. We propose a model whereby DNA shape features of stimulation-dependent genomic elements contribute to differences in the accessible chromatin landscape of alternatively activated macrophages on different genetic backgrounds that may contribute to phenotypic variations in immune responses.
Collapse
Affiliation(s)
- Mei San Tang
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016
| | - Emily R Miraldi
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229.,Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Natasha M Girgis
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016
| | - Richard A Bonneau
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY 10003.,Simons Center for Data Analysis, Simons Foundation, New York, NY 10011; and
| | - P'ng Loke
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016; .,Simons Center for Data Analysis, Simons Foundation, New York, NY 10011; and.,Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20814
| |
Collapse
|
34
|
Locati M, Curtale G, Mantovani A. Diversity, Mechanisms, and Significance of Macrophage Plasticity. ANNUAL REVIEW OF PATHOLOGY 2020; 15:123-147. [PMID: 31530089 PMCID: PMC7176483 DOI: 10.1146/annurev-pathmechdis-012418-012718] [Citation(s) in RCA: 1336] [Impact Index Per Article: 267.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Macrophages are a diverse set of cells present in all body compartments. This diversity is imprinted by their ontogenetic origin (embryonal versus adult bone marrow-derived cells); the organ context; by their activation or deactivation by various signals in the contexts of microbial invasion, tissue damage, and metabolic derangement; and by polarization of adaptive T cell responses. Classic adaptive responses of macrophages include tolerance, priming, and a wide spectrum of activation states, including M1, M2, or M2-like. Moreover, macrophages can retain long-term imprinting of microbial encounters (trained innate immunity). Single-cell analysis of mononuclear phagocytes in health and disease has added a new dimension to our understanding of the diversity of macrophage differentiation and activation. Epigenetic landscapes, transcription factors, and microRNA networks underlie the adaptability of macrophages to different environmental cues. Macrophage plasticity, an essential component of chronic inflammation, and its involvement in diverse human diseases, most notably cancer, is discussed here as a paradigm.
Collapse
Affiliation(s)
- Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, 20089 Milan, Italy
- Humanitas Clinical and Research Center, 20089 Milan, Italy;
| | - Graziella Curtale
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, 20089 Milan, Italy
- Humanitas Clinical and Research Center, 20089 Milan, Italy;
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, 20089 Milan, Italy;
- Humanitas University, 20090 Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| |
Collapse
|
35
|
Epigenetic Regulation of Inflammatory Cytokine-Induced Epithelial-To-Mesenchymal Cell Transition and Cancer Stem Cell Generation. Cells 2019; 8:cells8101143. [PMID: 31557902 PMCID: PMC6829508 DOI: 10.3390/cells8101143] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/20/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
The neoplastic transformation of normal to metastatic cancer cells is a complex multistep process involving the progressive accumulation of interacting genetic and epigenetic changes that alter gene function and affect cell physiology and homeostasis. Epigenetic changes including DNA methylation, histone modifications and changes in noncoding RNA expression, and deregulation of epigenetic processes can alter gene expression during the multistep process of carcinogenesis. Cancer progression and metastasis through an ‘invasion–metastasis cascade’ involving an epithelial-to-mesenchymal cell transition (EMT), the generation of cancer stem cells (CSCs), invasion of adjacent tissues, and dissemination are fueled by inflammation, which is considered a hallmark of cancer. Chronic inflammation is generated by inflammatory cytokines secreted by the tumor and the tumor-associated cells within the tumor microenvironment. Inflammatory cytokine signaling initiates signaling pathways leading to the activation of master transcription factors (TFs) such as Smads, STAT3, and NF-κB. Moreover, the same inflammatory responses also activate EMT-inducing TF (EMT-TF) families such as Snail, Twist, and Zeb, and epigenetic regulators including DNA and histone modifying enzymes and micoRNAs, through complex interconnected positive and negative feedback loops to regulate EMT and CSC generation. Here, we review the molecular regulatory feedback loops and networks involved in inflammatory cytokine-induced EMT and CSC generation.
Collapse
|
36
|
Clark AR, Ohlmeyer M. Protein phosphatase 2A as a therapeutic target in inflammation and neurodegeneration. Pharmacol Ther 2019; 201:181-201. [PMID: 31158394 PMCID: PMC6700395 DOI: 10.1016/j.pharmthera.2019.05.016] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric enzyme that catalyzes the selective removal of phosphate groups from protein serine and threonine residues. Emerging evidence suggests that it functions as a tumor suppressor by constraining phosphorylation-dependent signalling pathways that regulate cellular transformation and metastasis. Therefore, PP2A-activating drugs (PADs) are being actively sought and investigated as potential novel anti-cancer treatments. Here we explore the concept that PP2A also constrains inflammatory responses through its inhibitory effects on various signalling pathways, suggesting that PADs may be effective in the treatment of inflammation-mediated pathologies.
Collapse
Affiliation(s)
- Andrew R Clark
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
| | | |
Collapse
|
37
|
The Role of Genetic Profile in Functional Performance Adaptations to Exercise Training or Physical Activity: A Systematic Review of the Literature. J Aging Phys Act 2019; 27:594-616. [PMID: 30676214 DOI: 10.1123/japa.2018-0155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background: Variations in genotype may contribute to heterogeneity in functional adaptations to exercise. Methods: A systematic search of eight databases was conducted, and 9,696 citations were screened. Results: Eight citations from seven studies measuring 10 single-nucleotide polymorphisms and nine different functional performance test outcomes were included in the review. There was one observational study of physical activity and six experimental studies of aerobic or resistance training. The ACE (D) allele, ACTN3 (RR) genotype, UCP2 (GG) genotype, IL-6-174 (GG) genotype, TNF-α-308 (GG) genotype, and IL-10-1082 (GG) genotype all predicted significantly superior adaptations in at least one functional outcome in older men and women after prescribed exercise or in those with higher levels of physical activity. Conclusion: There is a small amount of evidence that older adults may have better functional outcomes after exercise/physical activity if they have specific alleles related to musculoskeletal function or inflammation. However, more robust trials are needed.
Collapse
|
38
|
Emerging Principles of Gene Expression Programs and Their Regulation. Mol Cell 2019; 71:389-397. [PMID: 30075140 DOI: 10.1016/j.molcel.2018.07.017] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/15/2018] [Accepted: 07/16/2018] [Indexed: 12/30/2022]
Abstract
Many mechanisms contribute to regulation of gene expression to ensure coordinated cellular behaviors and fate decisions. Transcriptional responses to external signals can consist of many hundreds of genes that can be parsed into different categories based on kinetics of induction, cell-type and signal specificity, and duration of the response. Here we discuss the structure of transcription programs and suggest a basic framework to categorize gene expression programs based on characteristics related to their control mechanisms. We also discuss possible evolutionary implications of this framework.
Collapse
|
39
|
Stubbs FE, Conway-Campbell BL, Lightman SL. Thirty years of neuroendocrinology: Technological advances pave the way for molecular discovery. J Neuroendocrinol 2019; 31:e12653. [PMID: 30362285 DOI: 10.1111/jne.12653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/16/2018] [Accepted: 10/21/2018] [Indexed: 12/12/2022]
Abstract
Since the 1950s, the systems level interactions between the hypothalamus, pituitary and end organs such as the adrenal, thyroid and gonads have been well known; however, it is only over the last three decades that advances in molecular biology and information technology have provided a tremendous expansion of knowledge at the molecular level. Neuroendocrinology has benefitted from developments in molecular genetics, epigenetics and epigenomics, and most recently optogenetics and pharmacogenetics. This has enabled a new understanding of gene regulation, transcription, translation and post-translational regulation, which should help direct the development of drugs to treat neuroendocrine-related diseases.
Collapse
Affiliation(s)
- Felicity E Stubbs
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK
| | - Becky L Conway-Campbell
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK
| | - Stafford L Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK
| |
Collapse
|
40
|
Rothenberg EV, Hosokawa H, Ungerbäck J. Mechanisms of Action of Hematopoietic Transcription Factor PU.1 in Initiation of T-Cell Development. Front Immunol 2019; 10:228. [PMID: 30842770 PMCID: PMC6391351 DOI: 10.3389/fimmu.2019.00228] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
PU.1 is an ETS-family transcription factor that plays a broad range of roles in hematopoiesis. A direct regulator of myeloid, dendritic-cell, and B cell functional programs, and a well-known antagonist of terminal erythroid cell differentiation, it is also expressed in the earliest stages of T-cell development of each cohort of intrathymic pro-T cells. Its expression in this context appears to give T-cell precursors initial, transient access to myeloid and dendritic cell developmental competence and therefore to represent a source of antagonism or delay of T-cell lineage commitment. However, it has remained uncertain until recently why T-cell development is also intensely dependent upon PU.1. Here, we review recent work that sheds light on the molecular biology of PU.1 action across the genome in pro-T cells and identifies the genes that depend on PU.1 for their correct regulation. This work indicates modes of chromatin engagement, pioneering, and cofactor recruitment (“coregulator theft”) by PU.1 as well as gene network interactions that not only affect specific target genes but also have system-wide regulatory consequences, amplifying the impact of PU.1 beyond its own direct binding targets. The genes directly regulated by PU.1 also suggest a far-reaching transformation of cell biology and signaling potential between the early stages of T-cell development when PU.1 is expressed and when it is silenced. These cell-biological functions can be important to distinguish fetal from adult T-cell development and have the potential to illuminate aspects of thymic function that have so far remained the most mysterious.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Hiroyuki Hosokawa
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Jonas Ungerbäck
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| |
Collapse
|
41
|
Hosokawa H, Ungerbäck J, Wang X, Matsumoto M, Nakayama KI, Cohen SM, Tanaka T, Rothenberg EV. Transcription Factor PU.1 Represses and Activates Gene Expression in Early T Cells by Redirecting Partner Transcription Factor Binding. Immunity 2019; 48:1119-1134.e7. [PMID: 29924977 DOI: 10.1016/j.immuni.2018.04.024] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/21/2018] [Accepted: 04/19/2018] [Indexed: 01/09/2023]
Abstract
Transcription factors normally regulate gene expression through their action at sites where they bind to DNA. However, the balance of activating and repressive functions that a transcription factor can mediate is not completely understood. Here, we showed that the transcription factor PU.1 regulated gene expression in early T cell development both by recruiting partner transcription factors to its own binding sites and by depleting them from the binding sites that they preferred when PU.1 was absent. The removal of partner factors Satb1 and Runx1 occurred primarily from sites where PU.1 itself did not bind. Genes linked to sites of partner factor "theft" were enriched for genes that PU.1 represses despite lack of binding, both in a model cell line system and in normal T cell development. Thus, system-level competitive recruitment dynamics permit PU.1 to affect gene expression both through its own target sites and through action at a distance.
Collapse
Affiliation(s)
- Hiroyuki Hosokawa
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jonas Ungerbäck
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA; Division of Molecular Hematology, Lund University, Sweden
| | - Xun Wang
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Masaki Matsumoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Japan
| | - Sarah M Cohen
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Tomoaki Tanaka
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Japan; AMED-CREST, AMED, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Ellen V Rothenberg
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
42
|
Ye Y, Liu Q, Zhang W, Mastwal S, Wang KH. Developmental Exposure to Psychostimulant Primes Activity-dependent Gene Induction in Frontal Cortex. Dev Neurobiol 2018; 79:96-108. [PMID: 30548567 DOI: 10.1002/dneu.22660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/08/2018] [Accepted: 11/12/2018] [Indexed: 11/11/2022]
Abstract
Perinatal neurodevelopment involves extensive formation of neural connections and onset of activity-dependent gene expression for synaptic function and plasticity. Exposure to psychostimulants at this stage imposes significant risks for developing cognitive and affective disorders later in life. However, how developmental exposure to psychostimulants may induce long-lasting molecular changes relevant to neural circuit function remains incompletely understood. In this study, we investigated the impacts of psychostimulant amphetamine on the activity-dependent induction of synaptic adaptor molecule Arc in the frontal cortex of neonatal mice. We found that transient exposure to amphetamine not only amplifies activity-dependent Arc mRNA expression immediately, but also potentiates subsequent induction of Arc mRNA in the absence of amphetamine. This priming effect is associated with a rapid and persistent increase in histone mono-methylation (H3K4me1), a marker for transcriptionally permissive chromatin, at the Arc locus, but not any long-lasting change in the phosphorylation of upstream transcription factor CREB. Furthermore, the increase in H3K4me1 at the Arc locus requires dopamine receptor signaling, and the priming of Arc expression correlates with the dopaminergic innervation pattern in the frontal cortex. Together, our results demonstrate that developmental exposure to psychostimulant amphetamine induces long-lasting chromatin changes and primes activity-dependent Arc gene induction. These findings reveal the molecular targets of psychostimulant during perinatal development that may contribute to long-term psychiatric risks.
Collapse
Affiliation(s)
- Yizhou Ye
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892
| | - Qing Liu
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892
| | - Wenyu Zhang
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892
| | - Surjeet Mastwal
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892
| | - Kuan Hong Wang
- Unit on Neural Circuits and Adaptive Behaviors, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892
| |
Collapse
|
43
|
Platanitis E, Decker T. Regulatory Networks Involving STATs, IRFs, and NFκB in Inflammation. Front Immunol 2018; 9:2542. [PMID: 30483250 PMCID: PMC6242948 DOI: 10.3389/fimmu.2018.02542] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/16/2018] [Indexed: 01/10/2023] Open
Abstract
Cells engaging in inflammation undergo drastic changes of their transcriptomes. In order to tailor these alterations in gene expression to the requirements of the inflammatory process, tight and coordinate regulation of gene expression by environmental cues, microbial or danger-associated molecules or cytokines, are mandatory. The transcriptional response is set off by signal-regulated transcription factors (SRTFs) at the receiving end of pathways originating at pattern recognition- and cytokine receptors. These interact with a genome that has been set for an appropriate response by prior activity of pioneer or lineage determining transcription factors (LDTFs). The same types of transcription factors are also critical determinants of the changes in chromatin landscapes and transcriptomes that specify potential consequences of inflammation: tissue repair, training, and tolerance. Here we focus on the role of three families of SRTFs in inflammation and its sequels: signal transducers and activators of transcription (STATs), interferon regulatory factors (IRFs), and nuclear factor κB (NFκB). We describe recent findings about their interactions and about their networking with LDTFs. Our aim is to provide a snapshot of a highly dynamic research area.
Collapse
Affiliation(s)
| | - Thomas Decker
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna, Austria
| |
Collapse
|
44
|
Ungerbäck J, Hosokawa H, Wang X, Strid T, Williams BA, Sigvardsson M, Rothenberg EV. Pioneering, chromatin remodeling, and epigenetic constraint in early T-cell gene regulation by SPI1 (PU.1). Genome Res 2018; 28:1508-1519. [PMID: 30171019 PMCID: PMC6169891 DOI: 10.1101/gr.231423.117] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 08/30/2018] [Indexed: 01/09/2023]
Abstract
SPI1 (also known as PU.1) is a dominant but transient regulator in early T-cell precursors and a potent transcriptional controller of developmentally important pro-T-cell genes. Before T-lineage commitment, open chromatin is frequently occupied by PU.1, and many PU.1 sites lose accessibility when PU.1 is later down-regulated. Pioneering activity of PU.1 was tested in this developmentally dynamic context by quantitating the relationships between PU.1 occupancy and site quality and accessibility as PU.1 levels naturally declined in pro-T-cell development and by using stage-specific gain- and loss-of-function perturbations to relate binding to effects on target genes. PU.1 could bind closed genomic sites, but rapidly opened many of them, despite the absence of its frequent collaborator, CEBPA. RUNX motifs and RUNX1 binding were often linked to PU.1 at open sites, but highly expressed PU.1 could bind its sites without RUNX1. The dynamic properties of PU.1 engagements implied that PU.1 binding affinity and concentration determine its occupancy choices, but with quantitative trade-offs for occupancy between site sequence quality and stage-dependent site accessibility in chromatin. At nonpromoter sites, PU.1 binding criteria were more stringent than at promoters, and PU.1 was also much more effective as a transcriptional regulator at nonpromoter sites where local chromatin accessibility depended on the presence of PU.1. Notably, closed chromatin presented a qualitative barrier to occupancy by the PU.1 DNA-binding domain alone. Thus, effective pioneering at closed chromatin sites also depends on requirements beyond site recognition, served by non-DNA-binding domains of PU.1.
Collapse
Affiliation(s)
- Jonas Ungerbäck
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA.,Division of Molecular Hematology, Lund University, SE-221 84 Lund, Sweden
| | - Hiroyuki Hosokawa
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Xun Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Tobias Strid
- Division of Molecular Hematology, Lund University, SE-221 84 Lund, Sweden
| | - Brian A Williams
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Mikael Sigvardsson
- Division of Molecular Hematology, Lund University, SE-221 84 Lund, Sweden.,Department of Clinical and Experimental Medicine, Linköping University, SE-581 83 Linköping, Sweden
| | - Ellen V Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| |
Collapse
|
45
|
PKCα-LSD1-NF-κB-Signaling Cascade Is Crucial for Epigenetic Control of the Inflammatory Response. Mol Cell 2018; 69:398-411.e6. [PMID: 29395062 DOI: 10.1016/j.molcel.2018.01.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/15/2017] [Accepted: 12/29/2017] [Indexed: 01/05/2023]
Abstract
The inflammatory response mediated by nuclear factor κB (NF-κB) signaling is essential for host defense against pathogens. Although the regulatory mechanism of NF-κB signaling has been well studied, the molecular basis for epigenetic regulation of the inflammatory response is poorly understood. Here we identify a new signaling axis of PKCα-LSD1-NF-κB, which is critical for activation and amplification of the inflammatory response. In response to excessive inflammatory stimuli, PKCα translocates to the nucleus and phosphorylates LSD1. LSD1 phosphorylation is required for p65 binding and facilitates p65 demethylation, leading to enhanced stability. In vivo genetic analysis using Lsd1SA/SA mice with ablation of LSD1 phosphorylation and chemical approaches in wild-type mice with inhibition of PKCα or LSD1 activity show attenuated sepsis-induced inflammatory lung injury and mortality. Together, we demonstrate that the PKCα-LSD1-NF-κB signaling cascade is crucial for epigenetic control of the inflammatory response, and targeting this signaling could be a powerful therapeutic strategy for systemic inflammatory diseases, including sepsis.
Collapse
|
46
|
Soldi M, Mari T, Nicosia L, Musiani D, Sigismondo G, Cuomo A, Pavesi G, Bonaldi T. Chromatin proteomics reveals novel combinatorial histone modification signatures that mark distinct subpopulations of macrophage enhancers. Nucleic Acids Res 2017; 45:12195-12213. [PMID: 28981749 PMCID: PMC5716071 DOI: 10.1093/nar/gkx821] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 09/06/2017] [Indexed: 01/09/2023] Open
Abstract
The integrated activity of cis-regulatory elements fine-tunes transcriptional programs of mammalian cells by recruiting cell type–specific as well as ubiquitous transcription factors (TFs). Despite their key role in modulating transcription, enhancers are still poorly characterized at the molecular level, and their limited DNA sequence conservation in evolution and variable distance from target genes make their unbiased identification challenging. The coexistence of high mono-methylation and low tri-methylation levels of lysine 4 of histone H3 is considered a signature of enhancers, but a comprehensive view of histone modifications associated to enhancers is still lacking. By combining chromatin immunoprecipitation (ChIP) with mass spectrometry, we investigated cis-regulatory regions in macrophages to comprehensively identify histone marks specifically associated with enhancers, and to profile their dynamics after transcriptional activation elicited by an inflammatory stimulation. The intersection of the proteomics data with ChIP-seq and RNA-seq analyses revealed the existence of novel subpopulations of enhancers, marked by specific histone modification signatures: specifically, H3K4me1/K36me2 marks transcribed enhancers, while H3K4me1/K36me3 and H3K4me1/K79me2 combinations mark distinct classes of intronic enhancers. Thus, our MS analysis of functionally distinct genomic regions revealed the combinatorial code of histone modifications, highlighting the potential of proteomics in addressing fundamental questions in epigenetics.
Collapse
Affiliation(s)
- Monica Soldi
- Department of Experimental Oncology, European Institute of Oncology, Milan 20139, Italy
| | - Tommaso Mari
- Department of Experimental Oncology, European Institute of Oncology, Milan 20139, Italy
| | - Luciano Nicosia
- Department of Experimental Oncology, European Institute of Oncology, Milan 20139, Italy
| | - Daniele Musiani
- Department of Experimental Oncology, European Institute of Oncology, Milan 20139, Italy
| | - Gianluca Sigismondo
- Department of Experimental Oncology, European Institute of Oncology, Milan 20139, Italy
| | - Alessandro Cuomo
- Department of Experimental Oncology, European Institute of Oncology, Milan 20139, Italy
| | - Giulio Pavesi
- Department of Biosciences, Milan University, Milan 20133, Italy
| | - Tiziana Bonaldi
- Department of Experimental Oncology, European Institute of Oncology, Milan 20139, Italy
| |
Collapse
|
47
|
Boniakowski AE, Kimball AS, Jacobs BN, Kunkel SL, Gallagher KA. Macrophage-Mediated Inflammation in Normal and Diabetic Wound Healing. THE JOURNAL OF IMMUNOLOGY 2017. [PMID: 28630109 DOI: 10.4049/jimmunol.1700223] [Citation(s) in RCA: 354] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The healing of cutaneous wounds is dependent on the progression through distinct, yet overlapping phases of wound healing, including hemostasis, inflammation, proliferation, and resolution/remodeling. The failure of these phases to occur in a timely, progressive fashion promotes pathologic wound healing. The macrophage (MΦ) has been demonstrated to play a critical role in the inflammatory phase of tissue repair, where its dynamic plasticity allows this cell to mediate both tissue-destructive and -reparative functions. The ability to understand and control both the initiation and the resolution of inflammation is critical for treating pathologic wound healing. There are now a host of studies demonstrating that metabolic and epigenetic regulation of gene transcription can influence MΦ plasticity in wounds. In this review, we highlight the molecular and epigenetic factors that influence MΦ polarization in both physiologic and pathologic wound healing, with particular attention to diabetic wounds.
Collapse
Affiliation(s)
- Anna E Boniakowski
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Andrew S Kimball
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI 48109; and
| | - Benjamin N Jacobs
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI 48109; and
| | - Steven L Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Katherine A Gallagher
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI 48109;
| |
Collapse
|
48
|
Miller GE, Borders AE, Crockett AH, Ross KM, Qadir S, Keenan-Devlin L, Leigh AK, Ham P, Ma J, Arevalo JM, Ernst LM, Cole SW. Maternal socioeconomic disadvantage is associated with transcriptional indications of greater immune activation and slower tissue maturation in placental biopsies and newborn cord blood. Brain Behav Immun 2017; 64:276-284. [PMID: 28434870 PMCID: PMC5493326 DOI: 10.1016/j.bbi.2017.04.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/17/2017] [Accepted: 04/18/2017] [Indexed: 12/13/2022] Open
Abstract
Children from economically disadvantaged families experience worse cognitive, psychiatric, and medical outcomes compared to more affluent youth. Preclinical models suggest some of the adverse influence of disadvantage could be transmitted during gestation via maternal immune activation, but this hypothesis has not been tested in humans. It also remains unclear whether prenatal interventions can mitigate such effects. To fill these gaps, we conducted two studies. Study 1 characterized the socioeconomic conditions of 79 women during pregnancy. At delivery, placenta biopsies and umbilical blood were collected for transcriptional profiling. Maternal disadvantage was associated with a transcriptional profile indicative of higher immune activation and slower fetal maturation, particularly in pathways related to brain, heart, and immune development. Cord blood cells of disadvantaged newborns also showed indications of immaturity, as reflected in down-regulation of pathways that coordinate myeloid cell development. These associations were independent of fetal sex, and characteristics of mothers (age, race, adiposity, diabetes, pre-eclampsia) and babies (delivery method, gestational age). Study 2 performed the same transcriptional analyses in specimens from 20 women participating in CenteringPregnancy, a group-based psychosocial intervention, and 20 women in traditional prenatal care. In both placenta biopsies and cord blood, women in CenteringPregnancy showed up-regulation of transcripts found in Study 1 to be most down-regulated in conjunction with disadvantage. Collectively, these results suggest socioeconomic disparities in placental biology are evident at birth, and provide clues about the mechanistic origins of health disparities. They also suggest the possibility that psychosocial interventions could have mitigating influences.
Collapse
Affiliation(s)
- Gregory E. Miller
- Department of Psychology and Institute for Policy Research, Northwestern University, Evanston IL
| | - Ann E. Borders
- Department of Obstetrics & Gynecology, NorthShore University Health System, University of Chicago Pritzker School of Medicine, Evanston IL
| | - Amy H. Crockett
- Department of Obstetrics & Gynecology, Greenville Hospital System University Medical Center, Greenville SC
| | - Kharah M. Ross
- Department of Psychology and Institute for Policy Research, Northwestern University, Evanston IL
| | - Sameen Qadir
- Department of Obstetrics & Gynecology, NorthShore University Health System, University of Chicago Pritzker School of Medicine, Evanston IL
| | - Lauren Keenan-Devlin
- Department of Obstetrics & Gynecology, NorthShore University Health System, University of Chicago Pritzker School of Medicine, Evanston IL
| | - Adam K. Leigh
- Department of Psychology and Institute for Policy Research, Northwestern University, Evanston IL
| | - Paula Ham
- Department of Psychology and Institute for Policy Research, Northwestern University, Evanston IL
| | - Jeffrey Ma
- Division of Hematology-Oncology, UCLA AIDS Institute, Molecular Biology Institute, Jonsson Comprehensive Cancer Center, Norman Cousins Center, UCLA School of Medicine, Los Angeles CA
| | - Jesusa M.G. Arevalo
- Division of Hematology-Oncology, UCLA AIDS Institute, Molecular Biology Institute, Jonsson Comprehensive Cancer Center, Norman Cousins Center, UCLA School of Medicine, Los Angeles CA
| | - Linda M. Ernst
- Department of Pathology, NorthShore University Health System, University of Chicago Pritzker School of Medicine, Evanston IL
| | - Steve W. Cole
- Division of Hematology-Oncology, UCLA AIDS Institute, Molecular Biology Institute, Jonsson Comprehensive Cancer Center, Norman Cousins Center, UCLA School of Medicine, Los Angeles CA
| |
Collapse
|
49
|
Kirov H, Schwarzer M, Neugebauer S, Faerber G, Diab M, Doenst T. Metabolomic profiling in patients undergoing Off-Pump or On-Pump coronary artery bypass surgery. BMC Cardiovasc Disord 2017; 17:93. [PMID: 28381258 PMCID: PMC5381030 DOI: 10.1186/s12872-017-0518-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/08/2017] [Indexed: 12/01/2022] Open
Abstract
Background Coronary artery bypass surgery can be performed without (Off-Pump) or with cardiopulmonary bypass (On-Pump). Extracorporeal circulation and cardioplegic arrest may cause alterations in the plasma metabolome. We assessed metabolomic changes in patients undergoing On-Pump or Off-Pump coronary artery bypass surgery. Methods We assessed five analyte classes (41 acylcarnitines, 14 amino acids, 92 glycerophospholipids, 15 sphingolipids, sugars, lactate) using a mass-spectrometry-based kit (Biocrates AbsoluteIDQ® p150) in paired arterial and coronary sinus blood obtained from 10 consecutive On-Pump and 10 Off-Pump patients. Cardioplegia for On-Pump was warm blood Calafiore. On-Pump outcomes were corrected for hemodilution through crystalloid priming. Results Demographic data were equal in both groups with normal ejection fraction, renal and liver function. Patients received 2.25 ± 0.64 bypass grafts. All postoperative courses were uneventful. Of 164 measured metabolites, only 13 (7.9%) were altered by cardiopulmonary bypass. We found more long-chain acylcarnitines Off-Pump and more short-chain acylcarnitines On-Pump. Glycerophospholipids showed lower concentrations On-Pump and arginine (as the only different amino acid) Off-Pump. Interestingly, plasma arginine (nitric oxide precursor) concentration at the end of surgery correlated inversely with postoperative vasopressor need (r = −0.7; p < 0.001). Assessing arterial/venous differences revealed phosphatidylcholine-production and acylcarnitine-consumption. These findings were unaffected by cardiopulmonary bypass, cardioplegia or temporary vessel occlusion during Off-Pump surgery. Conclusions Cardiopulmonary bypass and warm blood cardioplegia cause only minor changes to the metabolomic profile of patients undergoing coronary artery bypass surgery. The observed changes affected mainly acylcarnitines. In addition, there appears to be a relationship between arginine and vasopressor need after bypass surgery. Electronic supplementary material The online version of this article (doi:10.1186/s12872-017-0518-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- H Kirov
- Department of Cardiothoracic Surgery, Friedrich Schiller University Jena, University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - M Schwarzer
- Department of Cardiothoracic Surgery, Friedrich Schiller University Jena, University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - S Neugebauer
- Department of Clinical Chemistry and Laboratory Medicine, Friedrich-Schiller-University Jena, University Hospital, Jena, Germany.,Integrated Research and Treatment Center, Center for Sepsis Control and Care (CSCC), Jena, Germany
| | - G Faerber
- Department of Cardiothoracic Surgery, Friedrich Schiller University Jena, University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - M Diab
- Department of Cardiothoracic Surgery, Friedrich Schiller University Jena, University Hospital, Am Klinikum 1, 07747, Jena, Germany.,Department of Cardiothoracic Surgery, Cairo University, Cairo, Egypt
| | - T Doenst
- Department of Cardiothoracic Surgery, Friedrich Schiller University Jena, University Hospital, Am Klinikum 1, 07747, Jena, Germany.
| |
Collapse
|
50
|
Cheng CS, Behar MS, Suryawanshi GW, Feldman KE, Spreafico R, Hoffmann A. Iterative Modeling Reveals Evidence of Sequential Transcriptional Control Mechanisms. Cell Syst 2017; 4:330-343.e5. [PMID: 28237795 PMCID: PMC5434763 DOI: 10.1016/j.cels.2017.01.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 11/30/2016] [Accepted: 01/13/2017] [Indexed: 02/03/2023]
Abstract
Combinatorial control of gene expression is presumed to be mediated by molecular interactions between coincident transcription factors (TFs). While information on the genome-wide locations of TFs is available, the genes they regulate and whether they function combinatorially often remain open questions. Here, we developed a mechanistic, rather than statistical, modeling approach to elucidate TF control logic from gene expression data. Applying this approach to hundreds of genes in 85 datasets measuring the transcriptional responses of murine fibroblasts and macrophages to cytokines and pathogens, we found that stimulus-responsive TFs generally function sequentially in logical OR gates or singly. Logical AND gates were found between NF-κB-responsive mRNA synthesis and MAPKp38-responsive control of mRNA half-life, but not between temporally coincident TFs. Our analyses identified the functional target genes of each of the pathogen-responsive TFs and prompt a revision of the conceptual underpinnings of combinatorial control of gene expression to include sequentially acting molecular mechanisms that govern mRNA synthesis and decay.
Collapse
Affiliation(s)
- Christine S Cheng
- Signaling Systems Laboratory, San Diego Center for Systems Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Marcelo S Behar
- Signaling Systems Laboratory, San Diego Center for Systems Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Microbiology, Immunology, and Molecular Genetics, Institute for Quantitative and Computational Biosciences (QCBio) and Molecular Biology Institute (MBI), University of California, Los Angeles, Los Angeles, CA 90025, USA
| | - Gajendra W Suryawanshi
- Department of Microbiology, Immunology, and Molecular Genetics, Institute for Quantitative and Computational Biosciences (QCBio) and Molecular Biology Institute (MBI), University of California, Los Angeles, Los Angeles, CA 90025, USA
| | - Kristyn E Feldman
- Signaling Systems Laboratory, San Diego Center for Systems Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Roberto Spreafico
- Department of Microbiology, Immunology, and Molecular Genetics, Institute for Quantitative and Computational Biosciences (QCBio) and Molecular Biology Institute (MBI), University of California, Los Angeles, Los Angeles, CA 90025, USA
| | - Alexander Hoffmann
- Signaling Systems Laboratory, San Diego Center for Systems Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Microbiology, Immunology, and Molecular Genetics, Institute for Quantitative and Computational Biosciences (QCBio) and Molecular Biology Institute (MBI), University of California, Los Angeles, Los Angeles, CA 90025, USA.
| |
Collapse
|