1
|
Liu Y, Liu X, Wang J, Xie Y, Guo J, Liu Z, Li Y, Jiang B, Wang J. Single-cell sequencing of peripheral blood mononuclear cells reveals immune landscape of monkeypox patients with HIV. Emerg Microbes Infect 2025; 14:2459136. [PMID: 39868995 PMCID: PMC11809181 DOI: 10.1080/22221751.2025.2459136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/06/2024] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
The monkeypox (MPXV) outbreak in 2022 is more prevalent among individuals with human immunodeficiency virus (HIV). While it is plausible that HIV-induced immunosuppression could result in a more severe progression, the exact mechanisms remain undetermined. To better understand the immunopathology of MPXV in patients with and without HIV infection, we employed single-cell RNA sequencing (scRNA-seq) to analyse peripheral blood mononuclear cells (PBMCs) from six patients hospitalized for MPXV, three of whom had HIV infection (HIV antibody positive and HIV RNA level below the detection limit), and three patients only infected with MPXV (HIV-). We map the peripheral immune response in both the acute phase and the recovery period, showing the reconfiguration of peripheral immune cell phenotypes in acute stage compared with recovery stage, characterized by disturbed cell subsets and intense cell interactions mediated by monocytes and neutrophils. Importantly, we also found obviously dysregulated gene expression and cell subsets in HIV+ patients proposing mechanism underlying their serious condition. Our findings provide a comprehensive cell atlas of MPXV patients, shed light on the mechanisms underlying the severe disease progression and longer recovery time in HIV+ individuals.
Collapse
Affiliation(s)
- Yamin Liu
- Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin, People’s Republic of China
| | - Xinhua Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, People’s Republic of China
| | - Jingjing Wang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Ying Xie
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Jing Guo
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Zhiqiang Liu
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Ying Li
- Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin, People’s Republic of China
| | - Bei Jiang
- Tianjin Institute of Hepatology, Tianjin Second People's Hospital, Tianjin, People’s Republic of China
| | - Jingya Wang
- State Key Laboratory of Experimental Hematology, Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, People’s Republic of China
| |
Collapse
|
2
|
Toghani D, Gupte S, Zeng S, Mahammadov E, Crosse EI, Seyedhassantehrani N, Burns C, Gravano D, Radtke S, Kiem HP, Rodriguez S, Carlesso N, Pradeep A, Georgiades A, Lucas F, Wilson NK, Kinston SJ, Göttgens B, Zong L, Beerman I, Park B, Janssens DH, Jones D, Toghani A, Nerlov C, Pietras EM, Mesnieres M, Maes C, Kumanogoh A, Worzfeld T, Cheong JG, Josefowicz SZ, Kharchenko P, Scadden DT, Scialdone A, Spencer JA, Silberstein L. Niche-derived Semaphorin 4A safeguards functional identity of myeloid-biased hematopoietic stem cells. NATURE AGING 2025; 5:558-575. [PMID: 39881190 PMCID: PMC12025894 DOI: 10.1038/s43587-024-00798-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025]
Abstract
Somatic stem cell pools comprise diverse, highly specialized subsets whose individual contribution is critical for the overall regenerative function. In the bone marrow, myeloid-biased hematopoietic stem cells (myHSCs) are indispensable for replenishment of myeloid cells and platelets during inflammatory response but, at the same time, become irreversibly damaged during inflammation and aging. Here we identify an extrinsic factor, Semaphorin 4A (Sema4A), which non-cell-autonomously confers myHSC resilience to inflammatory stress. We show that, in the absence of Sema4A, myHSC inflammatory hyper-responsiveness in young mice drives excessive myHSC expansion, myeloid bias and profound loss of regenerative function with age. Mechanistically, Sema4A is mainly produced by neutrophils, signals via a cell surface receptor, Plexin D1, and safeguards the myHSC epigenetic state. Our study shows that, by selectively protecting a distinct stem cell subset, an extrinsic factor preserves functional diversity of somatic stem cell pool throughout organismal lifespan.
Collapse
Affiliation(s)
- Dorsa Toghani
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sanika Gupte
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sharon Zeng
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Elmir Mahammadov
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum Muenchen, Munich, Germany
| | - Edie I Crosse
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Christian Burns
- Department of Bioengineering, University of California, Merced, Merced, CA, USA
| | - David Gravano
- Department of Bioengineering, University of California, Merced, Merced, CA, USA
| | - Stefan Radtke
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hans-Peter Kiem
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sonia Rodriguez
- Department of Stem Cell Biology & Regenerative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Nadia Carlesso
- Department of Stem Cell Biology & Regenerative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Amogh Pradeep
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Alexis Georgiades
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Fabienne Lucas
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nicola K Wilson
- Department of Haematology, Jeffrey Cheah Biomedical Centre, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Sarah J Kinston
- Department of Haematology, Jeffrey Cheah Biomedical Centre, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Berthold Göttgens
- Department of Haematology, Jeffrey Cheah Biomedical Centre, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Le Zong
- Epigenetics and Stem Cell Aging Unit, National Institute of Aging, Baltimore, MD, USA
| | - Isabel Beerman
- Epigenetics and Stem Cell Aging Unit, National Institute of Aging, Baltimore, MD, USA
| | - Bongsoo Park
- Epigenetics and Stem Cell Aging Unit, National Institute of Aging, Baltimore, MD, USA
| | - Derek H Janssens
- Department of Epigenetics, Van Del Institute, Grand Rapids, MI, USA
| | - Daniel Jones
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ali Toghani
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Claus Nerlov
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Eric M Pietras
- Department of Medicine-Hematology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marion Mesnieres
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Christa Maes
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, University of Osaka, Osaka, Japan
| | - Thomas Worzfeld
- Faculty of Medicine, Institute of Pharmacology, University of Marburg, Marburg, Germany
| | - Jin-Gyu Cheong
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Steven Z Josefowicz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Peter Kharchenko
- Department of Stem Cell and Regenerative Biology, Harvard University, Boston, MA, USA
| | - David T Scadden
- Department of Stem Cell and Regenerative Biology, Harvard University, Boston, MA, USA
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum Muenchen, Munich, Germany
| | - Joel A Spencer
- Department of Bioengineering, University of California, Merced, Merced, CA, USA
| | - Lev Silberstein
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| |
Collapse
|
3
|
Willems SH, Qian S, Lång P, Overtoom BE, Alimostafazadeh S, Fuentes-Mateos R, Vasse GF, van der Veen TA, Vlasma J, de Jager MH, Guryev V, Fejer G, Andersson G, Melgert BN. TRAPping the effects of tobacco smoking: the regulation and function of Acp5 expression in lung macrophages. Am J Physiol Lung Cell Mol Physiol 2025; 328:L497-L511. [PMID: 39993028 DOI: 10.1152/ajplung.00157.2024] [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: 05/13/2024] [Revised: 09/02/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025] Open
Abstract
Tartrate-resistant acid phosphatase [TRAP, gene acid phosphatase 5 (Acp5; gene name for TRAP)] is highly expressed in alveolar macrophages with proposed roles in lung inflammation and lung fibrosis development. We previously showed that its expression and activity are higher in lung macrophages of smokers and patients with chronic obstructive pulmonary disease (COPD), suggesting involvement in smoke-induced lung damage. In this study, we explored the function of TRAP and regulation of its different mRNA transcripts (Acp5 201-206) in lung tissue exposed to cigarette smoke to elucidate its function in alveolar macrophages. In mice exposed to cigarette smoke or air for 4-6 wk, higher Acp5 mRNA expression in lung tissue after smoking was mainly driven by transcript Acp5-202, which originates from macrophages. The expression of Acp5-202 correlated with transcription factors previously found to drive proliferation of macrophages. Treating fetal liver progenitor-derived alveolar-like macrophages [Max Planck Institute (MPI; macrophages derived from fetal liver progenitors) macrophages] with cigarette smoke extract resulted in more proliferation compared with nontreated cells. In contrast, Acp5-deficient MPI macrophages and MPI macrophages treated with a TRAP inhibitor proliferated significantly less than control macrophages. Mechanistically, this lack of proliferation after TRAP inhibition was associated with higher presence of phosphorylated Beta-catenin (β-catenin; a signaling protein) compared with nontreated controls. Phosphorylation of β-catenin is known to mark it for ubiquitination and degradation by the proteasome, preventing its activity in promoting cell proliferation. In conclusion, our findings provide strong evidence for TRAP stimulating alveolar macrophage proliferation by dephosphorylating β-catenin. By driving proliferation, TRAP likely helps sustain alveolar macrophage populations during smoke exposure, either compensating for their loss due to smoking or increasing their numbers to better manage smoke-induced damage.NEW & NOTEWORTHY This study has uncovered that the enzyme tartrate-resistant acid phosphatase (TRAP) is crucial for alveolar macrophage proliferation through a β-catenin-dependent pathway. Importantly, TRAP influences this important ability of alveolar macrophages through the Acp5-202 mRNA transcript. The increase in TRAP expression following smoke exposure suggests that it plays a key role in promoting cell renewal, potentially helping to mitigate smoke-induced lung damage.
Collapse
Affiliation(s)
- Suzanne H Willems
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Shilei Qian
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Pernilla Lång
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Bjarne E Overtoom
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Sina Alimostafazadeh
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Rocío Fuentes-Mateos
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Gwenda F Vasse
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - T Anienke van der Veen
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jelmer Vlasma
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marina H de Jager
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Victor Guryev
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gyorgy Fejer
- School of Biomedical and Healthcare Sciences, Peninsula Schools of Medicine and Dentistry, University of Plymouth, Plymouth, United Kingdom
| | - Göran Andersson
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Barbro N Melgert
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
Kong Y, Yue M, Xu C, Zhang J, Hong H, Lu J, Wang Y, Zhang X, Chen Q, Yang C, Liu HF, Qin J, Zhou J, Lee NY, Lin B, Tian X, Freeman GJ, Xia Y. RGMb drives macrophage infiltration to aggravate kidney disease. Proc Natl Acad Sci U S A 2025; 122:e2418739122. [PMID: 40080642 PMCID: PMC11929492 DOI: 10.1073/pnas.2418739122] [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: 09/15/2024] [Accepted: 02/03/2025] [Indexed: 03/15/2025] Open
Abstract
The importance of macrophages in kidney diseases has been well established; however, the mechanisms underlying the infiltration of macrophages into injured kidneys are not well understood. RGMb is a member of the repulsive guidance molecule (RGM) family. RGMb can be expressed on the cell surface but a large portion of RGMb is localized intracellularly. Among various immune cell types, macrophages express the highest levels of RGMb, but the biological functions of RGMb in macrophages remain largely unknown. We find that RGMb promoted macrophage migration in vitro and that in vivo, RGMb enhanced infiltration of macrophages into injured kidneys and aggravated kidney inflammation and injury in mice. Mechanistically, RGMb bound to TAB1 inside the cell and facilitated the interaction between TRAF6 ubiquitin ligase and TAB1, thereby promoting TRAF6-mediated K63-linked polyubiquitination and phosphorylation of TAK1, followed by increased αTAT1 phosphorylation and α-tubulin acetylation. The resulting changes in the cytoskeleton promoted macrophage migration in vitro and in vivo. Deletion of Rgmb in macrophages markedly reduced TAK1 phosphorylation, αTAT1 phosphorylation, and α-tubulin acetylation and attenuated macrophage infiltration, renal inflammation, tubular injury, and interstitial fibrosis during kidney injury. Our results suggest that macrophage RGMb promotes kidney disease by increasing macrophage infiltration via the TRAF6-TAB1-TAK1/αTAT1/α-tubulin cascade.
Collapse
Affiliation(s)
- Yonglun Kong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen518057, China
| | - Ming Yue
- AIDS Institute and Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chunhua Xu
- Department of Nephrology, Longgang Central Hospital of Shenzhen, Shenzhen518116, China
| | - Jing Zhang
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Huiling Hong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiahuan Lu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yang Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyi Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Qiuju Chen
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chen Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital, Guangdong Medical University, Zhanjiang524023, China
| | - Hua-Feng Liu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Affiliated Hospital, Guangdong Medical University, Zhanjiang524023, China
| | - Jinzhong Qin
- The Key Laboratory of Model Animal for Disease Study of Ministry of Education, Model Animal Research Center, Nanjing University, Nanjing210061, China
| | - Jingying Zhou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Nam Y. Lee
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ85724
| | - Bin Lin
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Xiaoyu Tian
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Yin Xia
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen518057, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
5
|
Khalil A, Dinh T, Parks M, Obeng RC, Gryder B, Kresak A, Wang Y, Maltas J, Bedrock M, Wei X, Faber Z, Rahm M, Scott J, LaFramboise T, Wang Z, McFarland C. In Vivo Multiplexed Modeling Reveals Diverse Roles of the TBX2 Subfamily and Egr1 in Ras -Driven Lung Adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.15.642187. [PMID: 40166332 PMCID: PMC11956923 DOI: 10.1101/2025.03.15.642187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The TBX2 subfamily of T-box transcription factors (including Tbx2 , Tbx3 , Tbx4 , Tbx5 ) plays an essential role in lung development. Downregulation of these genes in human Lung adenocarcinoma (LUAD) suggests that these genes may be tumor suppressive, however because downregulation appears to occur primarily via epigenetic change, it remains unclear if these changes causally drive tumor progression or are merely the consequence of upstream events. Herein, we developed the first multiplexed mouse model to study the impact of TBX2 subfamily loss, alongside associated signaling genes Egr1 , Chd2 , Tnfaip3a , and Atf3 , in Ras -driven lung cancer. Using TuBa-seq, a high-throughput tumor-barcoding system, we quantified the growth effects of these knockouts during early and late tumorigenesis. Chd2 loss consistently suppressed tumor progression, while Tbx2 loss exhibited stage-dependent effects. Notably, Egr1 emerged as a potent tumor suppressor, with its knockout increasing tumor size (∼5x) at 20 weeks, surpassing Rb1 loss. Transcriptomic analyses of Egr1 -deficient tumors suggested immune dysregulation, including heightened inflammation and potential markers of T cell exhaustion in the tumor microenvironment. These findings indicate that Egr1 may play a role in suppressing tumor growth through modulating immune dynamics, offering new insights into the interplay between tumor progression and immune regulation in LUAD.
Collapse
|
6
|
Pietrogrande G, Shaker MR, Stednitz SJ, Soheilmoghaddam F, Aguado J, Morrison SD, Zambrano S, Tabassum T, Javed I, Cooper-White J, Davis TP, O'Brien TJ, Scott EK, Wolvetang EJ. Valproic acid-induced teratogenicity is driven by senescence and prevented by Rapamycin in human spinal cord and animal models. Mol Psychiatry 2025; 30:986-998. [PMID: 39227432 PMCID: PMC11835743 DOI: 10.1038/s41380-024-02732-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
Valproic acid (VPA) is an effective and widely used anti-seizure medication but is teratogenic when used during pregnancy, affecting brain and spinal cord development for reasons that remain largely unclear. Here we designed a genetic recombinase-based SOX10 reporter system in human pluripotent stem cells that enables tracking and lineage tracing of Neural Crest cells (NCCs) in a human organoid model of the developing neural tube. We found that VPA induces extensive cellular senescence and promotes mesenchymal differentiation of human NCCs. We next show that the clinically approved drug Rapamycin inhibits senescence and restores aberrant NCC differentiation trajectory after VPA exposure in human organoids and in developing zebrafish, highlighting the therapeutic promise of this approach. Finally, we identify the pioneer factor AP1 as a key element of this process. Collectively our data reveal cellular senescence as a central driver of VPA-associated neurodevelopmental teratogenicity and identifies a new pharmacological strategy for prevention. These results exemplify the power of genetically modified human stem cell-derived organoid models for drug discovery.
Collapse
Affiliation(s)
- Giovanni Pietrogrande
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia.
| | - Mohammed R Shaker
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Sarah J Stednitz
- Department of Anatomy & Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Farhad Soheilmoghaddam
- School of Chemical Engineering, University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Julio Aguado
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Sean D Morrison
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Samuel Zambrano
- School of Medicine, Vita-Salute San Raffaele University, Milan, 20132, Italy
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, 20132, Italy
| | - Tahmina Tabassum
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Ibrahim Javed
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Justin Cooper-White
- School of Chemical Engineering, University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Terence J O'Brien
- Department of Neuroscience, The Central Clinical School, Alfred Health, Monash University, Melbourne, VIC, Australia
- The Departments of Medicine and Neurology, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Ethan K Scott
- Department of Anatomy & Physiology, University of Melbourne, Parkville, VIC, Australia
- Queensland Brain Institute, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| | - Ernst J Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, QLD, 4072, Australia
| |
Collapse
|
7
|
Xue Z, Hu Q, Lu X, Wang R. Systematic dynamical analysis reveals the hierarchical hematopoietic differentiation. Phys Rev E 2025; 111:034406. [PMID: 40247488 DOI: 10.1103/physreve.111.034406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/27/2025] [Indexed: 04/19/2025]
Abstract
The differentiation of hematopoietic stem cells has been investigated through extensive experimental studies, establishing a paradigm for stem cell differentiation studies. However, a systematic dynamical analysis of the intricate lineage progression in hematopoiesis has yet to be fully elucidated. In this paper, we construct a dynamical model which allows us to identify seven essential cell states throughout the hierarchical process of hematopoietic differentiation. Starting from common myeloid progenitors, the model tracks the progression through granulocyte-monocyte progenitors and megakaryocyte-erythrocyte progenitors, ultimately giving rise to the generation of monocytes, granulocytes, erythrocytes, and megakaryocytes. By performing systematic perturbations and statistical analyses, we uncover the core networks associated with these cell states and the mechanisms underlying cell fate transitions. For a system of ordinary differential equations describing a known gene regulatory network, by performing random searches within a high-dimensional parameter space, we can create a mapping between specific parameter configurations and diverse cell fates, thus providing profound insights into the intricate processes underlying cellular differentiation. The model presented here provides a comprehensive description of the underlying dynamics and gives deeper insights into the intricate mechanisms of cellular fate determination during the hierarchical cascade of hematopoietic differentiation.
Collapse
Affiliation(s)
- Zhuozhen Xue
- Shanghai University, Department of Mathematics, Shanghai 200444, China
| | - Qing Hu
- Shanghai University, Department of Mathematics, Shanghai 200444, China
| | - Xiaoqi Lu
- Shanghai University, Department of Mathematics, Shanghai 200444, China
| | - Ruiqi Wang
- Shanghai University, Department of Mathematics, Shanghai 200444, China
- Shanghai University, Newtouch Center for Mathematics of Shanghai University, Shanghai 200444, China
| |
Collapse
|
8
|
Liu J, Wang L, Zhong W, Cai J, Sun Y, Li S, Li J, Liu Y, Xiong F. Single-Cell RNA Sequencing Reveals Peripheral Immune Cell Senescence and Inflammatory Phenotypes in Patients with Premature Ovarian Failure. J Inflamm Res 2025; 18:2699-2715. [PMID: 40026314 PMCID: PMC11871908 DOI: 10.2147/jir.s496130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/15/2025] [Indexed: 03/05/2025] Open
Abstract
Background Premature Ovarian Failure (POF) is a heterogeneous syndrome characterized by ovarian dysfunction, frequently associated with autoimmune factors. The interaction between peripheral and ovarian immune signals remains unclear. Recent advancements in single-cell technology provide a unique opportunity to examine the complex peripheral immune response in POF patients at the microstructural level. This study investigates the immune microenvironment's complexity through the interaction between peripheral and ovarian local immune responses. Methods Peripheral blood mononuclear cells (PBMCs) were isolated from three healthy individuals and four POF patients. Single-cell RNA sequencing (scRNA-seq) was used to delineate cell clusters and identify differentially expressed genes (DEGs). Enrichment, SCENIC, and pseudo-time analyses were utilized to explore cellular phenotype diversity, regulatory patterns, and evolutionary trajectories. A POF mouse model was used for validation. Results Seven clusters were identified and classified into two groups. POF patients exhibited increased proportions in T cells, NK cells, and B cells as well as upregulated IGLC2, GNLY, GZMB, FCGR3A, and CCL5 expressions compared to healthy controls. Monocytes, particularly non-classical monocytes, exhibited inflammatory phenotypes. CD8+ Effector T cells demonstrated increased cytotoxicity and TCR clonal expansion. The trajectory of CD8+ Effector T cells in POF patients involved the synchronous upregulation of cytotoxic-related genes and immune checkpoint molecules. Notably, CCL5, primarily produced by non-classical monocytes, emerged as a critical factor. Elevated levels of CCL5 in plasma and local ovaries, along with increased CD8+ T cell infiltration, suggested its potential role in chemotaxis and ovarian damage in POF. Validation in the POF mouse model further supported these findings. Conclusion In summary, this study provides in-depth insights into the immune landscape of POF, revealing distinct cell populations, pathways, and signaling networks linked to the disease. These findings enhance our understanding of POF's immunological mechanisms, contributing to the development of potential diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Jianan Liu
- Department of Medical Genetics/Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Li Wang
- Reproductive Medicine Department, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, People’s Republic of China
| | - Weijun Zhong
- Department of Medical Genetics/Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
| | - Jing Cai
- Reproductive Medicine Department, Dongguan Maternal and Children Health Hospital, Dongguan, Guangdong, People’s Republic of China
| | - Yan Sun
- Reproductive Medicine Department, Dongguan Maternal and Children Health Hospital, Dongguan, Guangdong, People’s Republic of China
| | - SongJun Li
- Reproductive Medicine Department, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, People’s Republic of China
| | - Jiayi Li
- The First Clinical Medical School, Nan Fang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yanhui Liu
- Reproductive Medicine Department, The Third Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, People’s Republic of China
| | - Fu Xiong
- Department of Medical Genetics/Experimental Education/Administration Center, School of Basic Medical Sciences, Southern Medical University, Guangzhou, People’s Republic of China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, Guangdong, People’s Republic of China
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
9
|
Zhao Y, Patel J, Fan J, Wang X, Chen L, Li Y, Luo Z. Integrated analysis reveals that EGR1 promotes epithelial IL33 production in T2 asthma. J Transl Med 2025; 23:203. [PMID: 39966984 PMCID: PMC11837401 DOI: 10.1186/s12967-025-06116-y] [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: 11/04/2024] [Accepted: 01/08/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Airway epithelial cells constitute the first line of defense against external noxious stimuli and play crucial roles in the release of epithelial inflammatory cytokines (IL33, IL25 and TSLP), initiating airway allergic inflammatory diseases such as asthma. IL33 plays critical physiological processes in T2-endotype asthma. However, the mechanisms by which allergen exposure triggers IL33 release from airway epithelial cells remain unclear. METHODS Integrated bioinformatic analysis and transcriptional analysis of bulk RNA-seq and single cell RNA-seq (scRNA-seq) data were used to identify core genes and determine the internal gene network associated with IL33. The expression of EGR1 was subsequently analyzed in vitro in the BEAS-2B cell line and in vivo in a house dust mite (HDM)-induced mouse asthma model. The functional experiments of EGR1 were investigated in vitro via siRNA knockdown and over-expressed plasmid. Chromatin immunoprecipitation (ChIP)-PCR and dual-luciferase reporter assay validation were subsequently performed to investigate the mechanisms by which EGR1 regulates IL33 secretion. RESULTS Bulk RNA-seq and scRNA-seq data identified EGR1 as an epithelial cell-derived gene implicated in IL33 expressions in asthma. The comprehensive analysis of multiple datasets indicated that the high EGR1 expression in epithelial cells may suggest a mechanistic basis of T2-endotype childhood asthma. Moreover, we verified that the expressions of EGR1 in airway epithelial cells were elevated both in vitro and in vivo asthma models. EGR1 regulated the production of IL33. Ultimately, ChIP and luciferase reporter assays confirmed that transcription factor EGR1 directly regulate the transcription of IL33 mRNA. CONCLUSIONS Our integrated bioinformatic analysis elucidated that EGR1 directly regulates the production of IL33 in T2-asthma and provide insights underlying the progression of asthma.
Collapse
Affiliation(s)
- Yan Zhao
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, China
| | - Jenil Patel
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston (UTHealth Houston) School of Public Health, Dallas, TX, USA
| | - Jinhua Fan
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyang Wang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Chen
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuanyuan Li
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Zhengxiu Luo
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
- China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing, China.
- Department of Respiratory Medicine, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Children's Hospital of Chongqing Medical University, No.136 Zhongshan 2nd Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
10
|
Sun B, Long Y, Xu G, Chen J, Wu G, Liu B, Gao Y. Acute hypoxia modulate macrophage phenotype accompanied with transcriptome re-programming and metabolic re-modeling. Front Immunol 2025; 16:1534009. [PMID: 40034701 PMCID: PMC11872928 DOI: 10.3389/fimmu.2025.1534009] [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: 11/25/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Macrophages, which tend to aggregate in the hypoxic regions of tissues, have a significant impact on disease progression and outcome because of their plastic responsiveness to hypoxia, particularly in the early stages. Understanding macrophages'participation in hypoxia-related disorders requires demonstrating the impact of acute hypoxia on their survival, phenotype, and function. Methods Here we conducted a systematic evaluation of macrophage responses to hypoxia over 24 and 48 h including cell growth and activity, inflamatory response, macrophage polarization and transcriptional and metabolic changes. Results We found that acute hypoxia suppresses macrophage proliferation and phagocytosis function with a parallel change of transcriptome re-programming and metabolic re-modeling. Although macrophages accumulate transcriptome heterogeneity based on oxygen concentration and culture period, genes involved in hypoxia response, chemotaxis, and glycolytic process were commonly altered during acute hypoxia. Furthermore, the pro-inflammatory response of macrophages was activated during acute hypoxia concomitantly with an enhanced anti-inflammatory regulatory mechanism characterized by increased M2 macrophage population and anti-inflammatory metabolite itaconic acid. Aside from increased glycolysis, the key intermediates in the pentose phosphate pathway significantly increased, such as fructose 1,6-bisphosphate (fold change: 7.8), 6-phosphogluconate (fold change: 6.1), and ribose 5-phosphate (fold change: 3.9), which indicated that the pentose phosphate pathway was an important compensatory metabolic regulation that rules for the response of macrophages to acute hypoxia. Discussion These findings highlight that acute hypoxia suppresses macrophage viability and phagocytosis, while acute hypoxia modifies the transcriptome and metabolome in specific inflammatory responses and metabolic pathways to facilitate the adaptation of macrophage in hypoxic conditions.
Collapse
Affiliation(s)
- Binda Sun
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
- Key Laboratory of High Altitude Medicine, Chinese People’s Liberation Army (PLA), Chongqing, China
| | - Yao Long
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
- Key Laboratory of High Altitude Medicine, Chinese People’s Liberation Army (PLA), Chongqing, China
| | - Gang Xu
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
- Key Laboratory of High Altitude Medicine, Chinese People’s Liberation Army (PLA), Chongqing, China
| | - Jian Chen
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
- Key Laboratory of High Altitude Medicine, Chinese People’s Liberation Army (PLA), Chongqing, China
| | - Gang Wu
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
- Key Laboratory of High Altitude Medicine, Chinese People’s Liberation Army (PLA), Chongqing, China
| | - Bao Liu
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
- Key Laboratory of High Altitude Medicine, Chinese People’s Liberation Army (PLA), Chongqing, China
- Department of High Altitude Physiology and Pathology, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuqi Gao
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
- Key Laboratory of High Altitude Medicine, Chinese People’s Liberation Army (PLA), Chongqing, China
| |
Collapse
|
11
|
Han Y, Liu C, Nie Y, Jiang X, An N, Qin Y, Ma Y, An Z, Zhao Y. Sex differences in the association between serum α-Klotho levels and hyperlipidemia: a cross-sectional study from NHANES 2013-2016. Sci Rep 2025; 15:4919. [PMID: 39929854 PMCID: PMC11811052 DOI: 10.1038/s41598-024-85018-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025] Open
Abstract
This study assessed the association between serum α-Klotho levels and hyperlipidemia. The aim was to ascertain the potential of serum α-Klotho levels as a predictive biomarker for hyperlipidemia. The research employed data from the National Health and Nutrition Examination Survey (NHANES) between 2013 and 2016. Weighted logistic regression analyses were employed to investigate the association between serum α-Klotho levels and hyperlipidemia. Restricted cubic spline (RCS) analyses were executed for males and females to scrutinize the non-linear correlation between α-Klotho levels and hyperlipidemia. Subsequently, piecewise logistic regression analysis was carried out based on the RCS findings. In females, the levels of α-Klotho were notably lower in those with hyperlipidemia in comparison to those without this condition (P < 0.05), No significant variation was demonstrated in α-Klotho levels between males with and without hyperlipidemia (P > 0.05). The participants were stratified by sex and subjected to analysis by logistic regression model. When α-Klotho was log2-transformed, it was significantly negatively associated with the risk of hyperlipidemia in females, even after adjusting for all of the covariates (OR 0.45, 95% CI 0.24-0.82), which was not observed in males (OR 1.14, 95% CI 0.63-2.06). The same results were observed in the third tertile of α-Klotho. Moreover, RCS analysis suggested a nonlinear correlation between serum α-Klotho levels and hyperlipidemia in females (P < 0.01). The inflection point of α-Klotho was found to be 1106.87 pg/mL. The piecewise logistic regression model revealed that when α-Klotho levels exceeded 1106.87 pg/mL, the link between α-Klotho and hyperlipidemia was no longer significant (P > 0.05). This investigation highlights the sex-based variation in the link between serum α-Klotho levels and hyperlipidemia. In females, α-Klotho exhibited negative association with hyperlipidemia, displaying a saturation effect. Serum α-Klotho emerges as a promising biological marker for the risk of hyperlipidemia among females.
Collapse
Affiliation(s)
- Yu Han
- Department of Pharmacy, Hebei Children's Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Chao Liu
- Department of Laboratory Animal Science, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
- Hebei Key Lab of Laboratory Animal Science, Shijiazhuang, Hebei, People's Republic of China
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Ying Nie
- College of Pharmacy, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Xijuan Jiang
- Department of Pharmacy, Hebei Children's Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Na An
- Department of Pharmacy, Hebei Children's Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Yabin Qin
- Department of Pharmacy, Hebei Children's Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Yinghua Ma
- Department of Pharmacy, Hebei Children's Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Zhihua An
- Department of Pharmacy, Hebei Children's Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Yile Zhao
- Department of Pharmacy, Hebei Children's Hospital, Shijiazhuang, Hebei, People's Republic of China.
| |
Collapse
|
12
|
Wu X, Pan T, Fang Z, Hui T, Yu X, Liu C, Guo Z, Liu C. Identification of EGR1 as a Key Diagnostic Biomarker in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) Through Machine Learning and Immune Analysis. J Inflamm Res 2025; 18:1639-1656. [PMID: 39925925 PMCID: PMC11806694 DOI: 10.2147/jir.s499396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/25/2025] [Indexed: 02/11/2025] Open
Abstract
Background Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), as a common chronic liver condition globally, is experiencing an increasing incidence rate which poses significant health risks. Despite this, the detailed mechanisms underlying the disease's onset and progression remain poorly understood. In this study, we aim to identify effective diagnostic biomarkers for MASLD using microarray data combined with machine learning techniques, which will aid in further understanding the pathogenesis of MASLD. Methods We collected six datasets from the Gene Expression Omnibus (GEO) database, using five of them as training sets and one as a validation set. We employed three machine learning methods-LASSO, SVM, and Random Forest (RF)-to identify hub genes associated with MASLD. These genes were further validated using the external dataset GSE164760. Additionally, functional enrichment analysis, immune infiltration analysis, and immune function analysis were conducted. A TF-miRNA-mRNA network was constructed, and single-cell RNA sequencing was used to determine the distribution of key genes within key cell clusters. Finally, the expression of the key genes was further validated using the palmitic acid-induced AML-12 cell line and the MCD mouse model. Results In this study, through differential gene expression (DEGs) analysis and machine learning techniques, we successfully identified 10 hub genes. Among these, the key gene EGR1 was validated and screened using an external dataset, with an area under the curve (AUC) of 0.882. Enrichment analyses and immune infiltration assessments revealed multiple pathways involving EGR1 in the pathogenesis and progression of MASLD, showing significant correlations with various immune cells. Furthermore, additional cellular experiments and animal model validations confirmed that the expression trends of EGR1 are highly consistent with our analytical findings. Conclusion Our research has confirmed EGR1 as a key gene in MASLD, providing novel insights into the disease's pathogenesis and identifying new therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Xuanlin Wu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Tao Pan
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Zhihao Fang
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Titi Hui
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Xiaoxiao Yu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Changxu Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Zihao Guo
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Chang Liu
- Department of General Surgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
13
|
King EM, Zhao Y, Moore CM, Steinhart B, Anderson KC, Vestal B, Moore PK, McManus SA, Evans CM, Mould KJ, Redente EF, McCubbrey AL, Janssen WJ. Gpnmb and Spp1 mark a conserved macrophage injury response masking fibrosis-specific programming in the lung. JCI Insight 2024; 9:e182700. [PMID: 39509324 PMCID: PMC11665561 DOI: 10.1172/jci.insight.182700] [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: 05/31/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
Macrophages are required for healthy repair of the lungs following injury, but they are also implicated in driving dysregulated repair with fibrosis. How these 2 distinct outcomes of lung injury are mediated by different macrophage subsets is unknown. To assess this, single-cell RNA-Seq was performed on lung macrophages isolated from mice treated with LPS or bleomycin. Macrophages were categorized based on anatomic location (airspace versus interstitium), developmental origin (embryonic versus recruited monocyte derived), time after inflammatory challenge, and injury model. Analysis of the integrated dataset revealed that macrophage subset clustering was driven by macrophage origin and tissue compartment rather than injury model. Gpnmb-expressing recruited macrophages that were enriched for genes typically associated with fibrosis were present in both injury models. Analogous GPNMB-expressing macrophages were identified in datasets from both fibrotic and nonfibrotic lung disease in humans. We conclude that this subset represents a conserved response to tissue injury and is not sufficient to drive fibrosis. Beyond this conserved response, we identified that recruited macrophages failed to gain resident-like programming during fibrotic repair. Overall, fibrotic versus nonfibrotic tissue repair is dictated by dynamic shifts in macrophage subset programming and persistence of recruited macrophages.
Collapse
Affiliation(s)
- Emily M. King
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Yifan Zhao
- Center for Genes, Environment, and Health, and
| | | | | | | | | | - Peter K. Moore
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Christopher M. Evans
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kara J. Mould
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Elizabeth F. Redente
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Alexandra L. McCubbrey
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - William J. Janssen
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
14
|
Venkatraman S, Balasubramanian B, Kongpracha P, Yangngam S, Chuangchot N, Khanaruksombat S, Thongchot S, Suntiparpluacha M, Myint KZ, Soodvilai S, Janvilisri T, Jirawatnotai S, Thuwajit P, Thuwajit C, Meller J, Chutipongtanate S, Tohtong R. Identification of Transcriptional Regulators of Immune Evasion Across Cancers: An Alternative Immunotherapeutic Strategy for Cholangiocarcinoma. Cancers (Basel) 2024; 16:4197. [PMID: 39766097 PMCID: PMC11674672 DOI: 10.3390/cancers16244197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/02/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Cancer immune evasion is a multifaceted process that synchronizes pro-tumoral immune infiltration, immunosuppressive inflammation, and inhibitory immune checkpoint expression (IC). Current immunotherapies combat this issue by reinstating immunosurveillance of tumors; however, it benefits a limited patient population. Thus, a more effective immunotherapeutic strategy is warranted to cater to specific patient populations. This investigation introduces a novel immunotherapeutic strategy via inhibition of master regulators of immune evasion (MR-IE). METHODS Samples of the TCGA Pan-Cancer Atlas transcriptomic data were subset and stratified based on IC and estimated immune cell infiltration. Transcriptomic analysis was conducted to unravel pathways associated with the immune evasion process. Transcription factor enrichment and survival analyses were conducted to identify and rank candidate MR-IEs per cancer type. RESULTS Inhibition of the top-ranking MR-IE candidate of cholangiocarcinoma (CCA), MYC, modulated the gene and protein expression of PD-L1. Moreover, pro-tumoral inflammatory markers, IFNA21 and CX3CL1, were downregulated, and anti-tumoral cytokines, IL-18 and IL-16, were upregulated. Lastly, MYC inhibition potentiated fourth-generation anti-folate receptor alpha (FRα) CAR-T cell therapy against CCA cells. CONCLUSIONS Cumulatively, this study highlights the promise of MR-IE inhibition as a novel potent immunotherapeutic strategy for the treatment of CCA and offers a candidate list of MR-IEs per cancer type for further validation.
Collapse
Affiliation(s)
- Simran Venkatraman
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.V.); (B.B.); (K.Z.M.); (T.J.)
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Brinda Balasubramanian
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.V.); (B.B.); (K.Z.M.); (T.J.)
- Translational Medical Sciences Unit, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Pornparn Kongpracha
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, CA 94158, USA;
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Supaporn Yangngam
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (S.Y.); (N.C.); (S.K.); (S.T.); (P.T.); (C.T.)
| | - Nisa Chuangchot
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (S.Y.); (N.C.); (S.K.); (S.T.); (P.T.); (C.T.)
- Siriraj Center of Research Excellence for Cancer Immunotherapy, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Suparada Khanaruksombat
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (S.Y.); (N.C.); (S.K.); (S.T.); (P.T.); (C.T.)
- Siriraj Center of Research Excellence for Cancer Immunotherapy, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Suyanee Thongchot
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (S.Y.); (N.C.); (S.K.); (S.T.); (P.T.); (C.T.)
- Siriraj Center of Research Excellence for Cancer Immunotherapy, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Monthira Suntiparpluacha
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (M.S.); (S.J.)
| | - Kyaw Zwar Myint
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.V.); (B.B.); (K.Z.M.); (T.J.)
| | - Sunhapas Soodvilai
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.V.); (B.B.); (K.Z.M.); (T.J.)
| | - Siwanon Jirawatnotai
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (M.S.); (S.J.)
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (S.Y.); (N.C.); (S.K.); (S.T.); (P.T.); (C.T.)
| | - Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; (S.Y.); (N.C.); (S.K.); (S.T.); (P.T.); (C.T.)
| | - Jarek Meller
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Somchai Chutipongtanate
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
| | - Rutaiwan Tohtong
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (S.V.); (B.B.); (K.Z.M.); (T.J.)
| |
Collapse
|
15
|
Li W, Ma X, Li X, Zhang X, Sun Y, Ning C, Zhang Q, Wang D, Tang H. Integrating Single-Cell RNA-Seq and ATAC-Seq Analysis Reveals Uterine Cell Heterogeneity and Regulatory Networks Linked to Pimpled Eggs in Chickens. Int J Mol Sci 2024; 25:13431. [PMID: 39769196 PMCID: PMC11679886 DOI: 10.3390/ijms252413431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/04/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Pimpled eggs have defective shells, which severely impacts hatching rates and transportation safety. In this study, we constructed single-cell resolution transcriptomic and chromatin accessibility maps from uterine tissues of chickens using single-cell RNA sequencing (scRNA-seq) and single-cell ATAC sequencing (scATAC-seq). We identified 11 major cell types and characterized their marker genes, along with specific transcription factors (TFs) that determine cell fate. CellChat analysis showed that fibroblasts had the most extensive intercellular communication network and that the chickens laying pimpled eggs had amplified immune-related signaling pathways. Differential expression and enrichment analyses indicated that inflammation in pimpled egg-laying chickens may lead to disruptions in their circadian rhythm and changes in the expression of ion transport-related genes, which negatively impacts eggshell quality. We then integrated TF analysis to construct a regulatory network involving TF-target gene-Gene Ontology associations related to pimpled eggs. We found that the transcription factors ATF3, ATF4, JUN, and FOS regulate uterine activities upstream, while the downregulation of ion pumps and genes associated with metal ion binding directly promotes the formation of pimpled eggs. Finally, by integrating the results of scRNA-seq and scATAC-seq, we identified a rare cell type-ionocytes. Our study constructed single-cell resolution transcriptomic and chromatin accessibility maps of chicken uterine tissue and explored the molecular regulatory mechanisms underlying pimpled egg formation. Our findings provide deeper insights into the structure and function of the chicken uterus, as well as the molecular mechanisms of eggshell formation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dan Wang
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China; (W.L.); (X.M.); (X.L.); (X.Z.); (Y.S.); (C.N.); (Q.Z.)
| | - Hui Tang
- Shandong Provincial Key Laboratory for Livestock Germplasm Innovation & Utilization, College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Taian 271018, China; (W.L.); (X.M.); (X.L.); (X.Z.); (Y.S.); (C.N.); (Q.Z.)
| |
Collapse
|
16
|
Zhang C, Lv P, Liang Q, Zhou J, Wu B, Xu W. Conditioned Medium Derived From Human Dental Follicle Mesenchymal Stem Cells Alleviates Macrophage Proinflammatory Responses Through MAPK-ERK-EGR1 Axis. Stem Cells Int 2024; 2024:5514771. [PMID: 39650749 PMCID: PMC11623994 DOI: 10.1155/sci/5514771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 10/09/2024] [Accepted: 11/02/2024] [Indexed: 12/11/2024] Open
Abstract
The regulation of macrophage polarization by mesenchymal stem cells (MSCs) is a prominent area of research but faces challenges due to limited MSC sources and incomplete understanding of underlying mechanisms. We sought to identify an accessible MSC source and investigate how MSCs regulate macrophage polarization using high-throughput sequencing. We isolated dental follicle MSCs from discarded human third molar dental follicles and cocultured them with THP-1-derived macrophages in the conditioned medium. Transcriptome sequencing identified differentially expressed genes (DEGs) in macrophages, integrating with multiomics database analysis to uncover polarization mechanisms. Our findings demonstrated successful MSC extraction from dental follicles, with the conditioned medium suppressing proinflammatory macrophage functions and influencing macrophage subtyping. MSCs, through paracrine signaling, activated the mitogen-activated protein kinase (MAPK) pathway, leading to extracellular regulated protein kinases (ERK)1/2 phosphorylation and upregulation of early growth response 1 (EGR1) protein. Elevated EGR1 levels inhibited inflammatory gene expression, inhibiting the pro-inflammatory immunoregulatory function of macrophages in inflammatory states. This study provides an efficient method for in vitro macrophage polarization identification. It offers insights into MSC-regulated polarization mechanisms, with potential clinical implications for anti-inflammatory therapy and immune regulation.
Collapse
Affiliation(s)
- Chuhan Zhang
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangzhou, China
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen, China
| | - Peiyi Lv
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangzhou, China
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen, China
| | - Qiuying Liang
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangzhou, China
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen, China
| | - Jian Zhou
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Buling Wu
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangzhou, China
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen, China
| | - Wenan Xu
- Shenzhen Clinical College of Stomatology, School of Stomatology, Southern Medical University, Guangzhou, China
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen, China
| |
Collapse
|
17
|
Zerad L, Gacem N, Gayda F, Day L, Sinigaglia K, Richard L, Parisot M, Cagnard N, Mathis S, Bole-Feysot C, O’Connell MA, Pingault V, Dambroise E, Keegan LP, Vallat JM, Bondurand N. Overexpression of Egr1 Transcription Regulator Contributes to Schwann Cell Differentiation Defects in Neural Crest-Specific Adar1 Knockout Mice. Cells 2024; 13:1952. [PMID: 39682701 PMCID: PMC11639873 DOI: 10.3390/cells13231952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/05/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Adenosine deaminase acting on RNA 1 (ADAR1) is the principal enzyme for the adenosine-to-inosine RNA editing that prevents the aberrant activation of cytosolic nucleic acid sensors by endogenous double stranded RNAs and the activation of interferon-stimulated genes. In mice, the conditional neural crest deletion of Adar1 reduces the survival of melanocytes and alters the differentiation of Schwann cells that fail to myelinate nerve fibers in the peripheral nervous system. These myelination defects are partially rescued upon the concomitant removal of the Mda5 antiviral dsRNA sensor in vitro, suggesting implication of the Mda5/Mavs pathway and downstream effectors in the genesis of Adar1 mutant phenotypes. By analyzing RNA-Seq data from the sciatic nerves of mouse pups after conditional neural crest deletion of Adar1 (Adar1cKO), we here identified the transcription factors deregulated in Adar1cKO mutants compared to the controls. Through Adar1;Mavs and Adar1cKO;Egr1 double-mutant mouse rescue analyses, we then highlighted that the aberrant activation of the Mavs adapter protein and overexpression of the early growth response 1 (EGR1) transcription factor contribute to the Adar1 deletion associated defects in Schwann cell development in vivo. In silico and in vitro gene regulation studies additionally suggested that EGR1 might mediate this inhibitory effect through the aberrant regulation of EGR2-regulated myelin genes. We thus demonstrate the role of the Mda5/Mavs pathway, but also that of the Schwann cell transcription factors in Adar1-associated peripheral myelination defects.
Collapse
Affiliation(s)
- Lisa Zerad
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| | - Nadjet Gacem
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| | - Fanny Gayda
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| | - Lucie Day
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| | - Ketty Sinigaglia
- Central European Institute for Technology, Masaryk University (CEITEC MU), Kamenice 735/5, 625 00 Brno, Czech Republic; (K.S.); (M.A.O.); (L.P.K.)
| | - Laurence Richard
- Department of Neurology, Centre de Reference “Neuropathies Périphériques Rares”, CHU Limoges, 87000 Limoges, France; (L.R.); (J.M.V.)
| | - Melanie Parisot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UAR3633, Paris Descartes Sorbonne Paris Cite University, 75015 Paris, France; (M.P.); (C.B.-F.)
| | - Nicolas Cagnard
- Bioinformatics Platform, Imagine Institute, INSERM UMR 1163, 75015 Paris, France;
| | - Stephane Mathis
- Department of Neurology (Nerve-Muscle Unit) and ‘Grand Sud-Ouest’ National Reference Center for Neuromuscular Disorders, CHU Bordeaux, Pellegrin Hospital, 33000 Bordeaux, France;
| | - Christine Bole-Feysot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/CNRS UAR3633, Paris Descartes Sorbonne Paris Cite University, 75015 Paris, France; (M.P.); (C.B.-F.)
| | - Mary A. O’Connell
- Central European Institute for Technology, Masaryk University (CEITEC MU), Kamenice 735/5, 625 00 Brno, Czech Republic; (K.S.); (M.A.O.); (L.P.K.)
| | - Veronique Pingault
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| | - Emilie Dambroise
- Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France;
| | - Liam P. Keegan
- Central European Institute for Technology, Masaryk University (CEITEC MU), Kamenice 735/5, 625 00 Brno, Czech Republic; (K.S.); (M.A.O.); (L.P.K.)
| | - Jean Michel Vallat
- Department of Neurology, Centre de Reference “Neuropathies Périphériques Rares”, CHU Limoges, 87000 Limoges, France; (L.R.); (J.M.V.)
| | - Nadege Bondurand
- Laboratory of Embryology and Genetics of Human Malformations, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 24 Boulevard du Montparnasse, 75015 Paris, France; (L.Z.); (N.G.); (F.G.); (L.D.); (V.P.)
| |
Collapse
|
18
|
Wang Z, Tan W, Li B, Chen J, Zhu J, Xu F, Tang F, Yoshida S, Zhou Y. LncRNA-MM2P regulates retinal neovascularization through M2 macrophage polarization. Exp Eye Res 2024; 248:110072. [PMID: 39241859 DOI: 10.1016/j.exer.2024.110072] [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: 04/13/2024] [Revised: 07/19/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
The study aims to investigate the effects and potential mechanisms of lncRNA-MM2P on retinal neovascularization in a mouse model of oxygen-induced retinopathy (OIR). The OIR model was established in C57BL/6J mice. RAW264.7 cell line and bone marrow-derived macrophages (BMDMs) from mice were used for in vitro studies. RT-qPCR was used to analyze the expressions of lncRNA and mRNAs. The protein expression levels were determined by western blotting. The size of avascular areas and neovascular tufts were assessed based on isolectin B4 immunofluorescence staining images. The human retinal endothelial cells (HRECs) were used to evaluate the proliferation, migration, and tube formation of endothelial cells. The expression of lncRNA-MM2P was significantly upregulated from P17 to P25 in OIR retinas. Knockdown of lncRNA-MM2P levels in vivo led to a significant reduction in the neovascular tufts and avascular areas in the retinas of OIR mice. Knockdown of lncRNA-MM2P levels in vitro suppressed the expression of M2 markers in macrophages. Moreover, we found a significant inhibition of avascular areas and neovascular tufts in OIR mice injected intravitreally with M2 macrophages treated by shRNA-MM2P. The cellular functions of proliferation, migration, and tube formation were significantly attenuated in HRECs cultured with a supernatant of shRNA-MM2P-treated M2 macrophages. Our results indicate that lncRNA-MM2P regulates retinal neovascularization by inducing M2 polarization of macrophages in OIR mice. Therefore, lncRNA-MM2P may be a potential molecular target for immunoregulation of retinal neovascularization.
Collapse
Affiliation(s)
- Zicong Wang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Wei Tan
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Bingyan Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Junyu Chen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Junye Zhu
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Fan Xu
- Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health, Nanning, Guangxi, 530021, China
| | - Fen Tang
- Department of Ophthalmology, The People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health, Nanning, Guangxi, 530021, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Fukuoka, 830-0011, Japan
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
19
|
Gabriel AAG, Racle J, Falquet M, Jandus C, Gfeller D. Robust estimation of cancer and immune cell-type proportions from bulk tumor ATAC-Seq data. eLife 2024; 13:RP94833. [PMID: 39383060 PMCID: PMC11464006 DOI: 10.7554/elife.94833] [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] [Indexed: 10/11/2024] Open
Abstract
Assay for Transposase-Accessible Chromatin sequencing (ATAC-Seq) is a widely used technique to explore gene regulatory mechanisms. For most ATAC-Seq data from healthy and diseased tissues such as tumors, chromatin accessibility measurement represents a mixed signal from multiple cell types. In this work, we derive reliable chromatin accessibility marker peaks and reference profiles for most non-malignant cell types frequently observed in the microenvironment of human tumors. We then integrate these data into the EPIC deconvolution framework (Racle et al., 2017) to quantify cell-type heterogeneity in bulk ATAC-Seq data. Our EPIC-ATAC tool accurately predicts non-malignant and malignant cell fractions in tumor samples. When applied to a human breast cancer cohort, EPIC-ATAC accurately infers the immune contexture of the main breast cancer subtypes.
Collapse
Affiliation(s)
- Aurélie Anne-Gaëlle Gabriel
- Department of Oncology, Ludwig Institute for Cancer Research, University of LausanneLausanneSwitzerland
- Agora Cancer Research CenterLausanneSwitzerland
- Swiss Cancer Center Leman (SCCL)GenevaSwitzerland
- Swiss Institute of Bioinformatics (SIB)LausanneSwitzerland
| | - Julien Racle
- Department of Oncology, Ludwig Institute for Cancer Research, University of LausanneLausanneSwitzerland
- Agora Cancer Research CenterLausanneSwitzerland
- Swiss Cancer Center Leman (SCCL)GenevaSwitzerland
- Swiss Institute of Bioinformatics (SIB)LausanneSwitzerland
| | - Maryline Falquet
- Swiss Cancer Center Leman (SCCL)GenevaSwitzerland
- Ludwig Institute for Cancer Research, Lausanne BranchLausanneSwitzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of GenevaGenevaSwitzerland
- Geneva Center for Inflammation ResearchGenevaSwitzerland
| | - Camilla Jandus
- Swiss Cancer Center Leman (SCCL)GenevaSwitzerland
- Ludwig Institute for Cancer Research, Lausanne BranchLausanneSwitzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of GenevaGenevaSwitzerland
- Geneva Center for Inflammation ResearchGenevaSwitzerland
| | - David Gfeller
- Department of Oncology, Ludwig Institute for Cancer Research, University of LausanneLausanneSwitzerland
- Agora Cancer Research CenterLausanneSwitzerland
- Swiss Cancer Center Leman (SCCL)GenevaSwitzerland
- Swiss Institute of Bioinformatics (SIB)LausanneSwitzerland
| |
Collapse
|
20
|
Yaman E, Heyer N, de Paiva CS, Stepp MA, Pflugfelder SC, Alam J. Mouse Corneal Immune Cell Heterogeneity Revealed by Single-Cell RNA Sequencing. Invest Ophthalmol Vis Sci 2024; 65:29. [PMID: 39432400 PMCID: PMC11500044 DOI: 10.1167/iovs.65.12.29] [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: 05/01/2024] [Accepted: 10/03/2024] [Indexed: 10/23/2024] Open
Abstract
Purpose This study aimed to define the heterogeneity, spatial localization, and functional roles of immune cells in the mouse cornea using single-cell RNA sequencing (scRNA-seq) and immunofluorescent staining. Methods Enriched mouse corneal immune cells (C57BL/6 strain, age 16-20 weeks) underwent single-cell RNA sequencing library preparation, sequencing, and analysis with Seurat, Monocle 3, and CellChat packages in R. Pathway analysis used Qiagen Ingenuity Pathway Analysis software. Immunostaining confirmed cell distribution. Results We identified 14 distinct immune cell clusters (56% myeloid and 44% lymphoid). Myeloid populations included resident macrophages, conventional dendritic cells (cDC2s), Langerhans cells, neutrophils, monocytes, and mast cells. Additionally, lymphocyte subsets (B, CD8, CD4, γδT, natural killer, natural killer T, and group 2 innate lymphoid cells) were found. We also found three new subtypes of resident macrophages in the cornea. Trajectory analysis suggested a differentiation pathway from monocytes to conventional dendritic cells, resident macrophages, and LCs. Intercellular communication network analysis using cord diagram identified amyloid precursor protein, chemokine (C-C motif) ligands (2, 3, 4, 6, 7, 9, and 12), Cxcl2, Mif, Tnf, Tgfb1, Igf1, and Il10 as prominent ligands involved in these interactions. Sexually dimorphic gene expression patterns were observed, with male myeloid cells expressing genes linked to immune regulation (Egr1, Foxp1, Mrc1, and Il1rn) and females showing higher expression of antigen presentation genes (Clic1, Psmb8, and Psmb9). Finally, immunostaining confirmed the spatial distribution of these cell populations within the cornea. Conclusions This study unveils a diverse immune landscape in the mouse cornea, with evidence for cell differentiation and sex-based differences. Immunostaining validates the spatial distribution of these populations, furthering our knowledge of corneal immune function.
Collapse
Affiliation(s)
- Ebru Yaman
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Nicole Heyer
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Cintia S. de Paiva
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Mary Ann Stepp
- Departments of Anatomy, Regenerative Biology and Ophthalmology, The George Washington University Medical School and Health Sciences, Washington, DC, United States
| | - Stephen C. Pflugfelder
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Jehan Alam
- Ocular Surface Center, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
21
|
Li R, Wei R, Liu C, Zhang K, He S, Liu Z, Huang J, Tang Y, An Q, Lin L, Gan L, Zhao L, Zou X, Wang F, Ping Y, Ma Q. Heme oxygenase 1-mediated ferroptosis in Kupffer cells initiates liver injury during heat stroke. Acta Pharm Sin B 2024; 14:3983-4000. [PMID: 39309491 PMCID: PMC11413699 DOI: 10.1016/j.apsb.2024.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 09/25/2024] Open
Abstract
With the escalating prevalence of global heat waves, heat stroke has become a prominent health concern, leading to substantial liver damage. Unlike other forms of liver injury, heat stroke-induced damage is characterized by heat cytotoxicity and heightened inflammation, directly contributing to elevated mortality rates. While clinical assessments have identified elevated bilirubin levels as indicative of Kupffer cell dysfunction, their specific correlation with heat stroke liver injury remains unclear. Our hypothesis proposes the involvement of Kupffer cell ferroptosis during heat stroke, initiating IL-1β-mediated inflammation. Using single-cell RNA sequencing of murine macrophages, a distinct and highly susceptible Kupffer cell subtype, Clec4F+/CD206+, emerged, with heme oxygenase 1 (HMOX-1) playing a pivotal role. Mechanistically, heat-induced HMOX-1, regulated by early growth response factor 1, mediated ferroptosis in Kupffer cells, specifically in the Clec4F+/CD206+ subtype (KC2), activating phosphatidylinositol 4-kinase beta and promoting PI4P production. This cascade triggered NLRP3 inflammasome activation and maturation of IL-1β. These findings underscore the critical role of targeted therapy against HMOX-1 in ferroptosis within Kupffer cells, particularly in Clec4F+/CD206+ KCs. Such an approach has the potential to mitigate inflammation and alleviate acute liver injury in the context of heat stroke, offering a promising avenue for future therapeutic interventions.
Collapse
Affiliation(s)
- Ru Li
- The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, China
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510000, China
| | - Riqing Wei
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510000, China
| | - Chenxin Liu
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510000, China
| | - Keying Zhang
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510000, China
| | - Sixiao He
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510000, China
| | - Zhifeng Liu
- Medical Critical Care Medicine, General Hospital of Southern Theatre Command of PLA, Guangzhou 510000, China
- Guangdong Branch Center, National Clinical Research Center for Geriatric Diseases (Chinese PLA General Hospital), Guangzhou 510000, China
| | - Junhao Huang
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510000, China
| | - Youyong Tang
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510000, China
| | - Qiyuan An
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510000, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Lishe Gan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Liying Zhao
- Department of General Surgery, Nanfang Hospital, the First School of Clinical Medicine, Southern Medical University, Guangzhou 510000, China
| | - Xiaoming Zou
- The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, China
| | - Fudi Wang
- The Fourth Affiliated Hospital, the First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Cancer Center, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310000, China
- The First Affiliated Hospital, the Second Affiliated Hospital, Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang 421200, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 310000, China
| | - Qiang Ma
- Department of Biopharmaceutics, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510000, China
- Guangdong Provincial Key Laboratory of Immune Regulation and Immunotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510000, China
| |
Collapse
|
22
|
Lahnsteiner A, Ellmer V, Oberlercher A, Liutkeviciute Z, Schönauer E, Paulweber B, Aigner E, Risch A. G-quadruplex forming regions in GCK and TM6SF2 are targets for differential DNA methylation in metabolic disease and hepatocellular carcinoma patients. Sci Rep 2024; 14:20215. [PMID: 39215018 PMCID: PMC11364803 DOI: 10.1038/s41598-024-70749-0] [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: 03/16/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
The alarming increase in global rates of metabolic diseases (MetDs) and their association with cancer risk renders them a considerable burden on our society. The interplay of environmental and genetic factors in causing MetDs may be reflected in DNA methylation patterns, particularly at non-canonical (non-B) DNA structures, such as G-quadruplexes (G4s) or R-loops. To gain insight into the mechanisms of MetD progression, we focused on DNA methylation and functional analyses on intragenic regions of two MetD risk genes, the glucokinase (GCK) exon 7 and the transmembrane 6 superfamily 2 (TM6SF2) intron 2-exon 3 boundary, which harbor non-B DNA motifs for G4s and R-loops.Pyrosequencing of 148 blood samples from a nested cohort study revealed significant differential methylation in GCK and TM6SF2 in MetD patients versus healthy controls. Furthermore, these regions harbor hypervariable and differentially methylated CpGs also in hepatocellular carcinoma versus normal tissue samples from The Cancer Genome Atlas (TCGA). Permanganate/S1 nuclease footprinting with direct adapter ligation (PDAL-Seq), native polyacrylamide DNA gel electrophoresis and circular dichroism (CD) spectroscopy revealed the formation of G4 structures in these regions and demonstrated that their topology and stability is affected by DNA methylation. Detailed analyses including histone marks, chromatin conformation capture data, and luciferase reporter assays, highlighted the cell-type specific regulatory function of the target regions. Based on our analyses, we hypothesize that changes in DNA methylation lead to topological changes, especially in GCK exon 7, and cause the activation of alternative regulatory elements or potentially play a role in alternative splicing.Our analyses provide a new view on the mechanisms underlying the progression of MetDs and their link to hepatocellular carcinomas, unveiling non-B DNA structures as important key players already in early disease stages.
Collapse
Affiliation(s)
- Angelika Lahnsteiner
- Division of Cancer (Epi-)Genetics, Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology (CTBI), Paris Lodron University Salzburg, Hellbrunnerstraße 34, 5020, Salzburg, Austria.
- Cancer Cluster Salzburg, Salzburg, Austria.
| | - Victoria Ellmer
- Division of Cancer (Epi-)Genetics, Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology (CTBI), Paris Lodron University Salzburg, Hellbrunnerstraße 34, 5020, Salzburg, Austria
| | - Anna Oberlercher
- Division of Cancer (Epi-)Genetics, Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology (CTBI), Paris Lodron University Salzburg, Hellbrunnerstraße 34, 5020, Salzburg, Austria
| | - Zita Liutkeviciute
- Division of Cancer (Epi-)Genetics, Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology (CTBI), Paris Lodron University Salzburg, Hellbrunnerstraße 34, 5020, Salzburg, Austria
| | - Esther Schönauer
- Division of Structural Biology, Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology (CTBI), Paris Lodron University Salzburg, Salzburg, Austria
| | - Bernhard Paulweber
- First Department of Medicine, University Clinic Salzburg, Salzburg, Austria
| | - Elmar Aigner
- First Department of Medicine, University Clinic Salzburg, Salzburg, Austria
- Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Angela Risch
- Division of Cancer (Epi-)Genetics, Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology (CTBI), Paris Lodron University Salzburg, Hellbrunnerstraße 34, 5020, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
23
|
Yeo H, Ahn SS, Ou S, Yun SJ, Lim Y, Koh D, Lee YH, Shin SY. The EGR1-Artemin Axis in Keratinocytes Enhances the Innervation of Epidermal Sensory Neurons during Skin Inflammation Induced by House Dust Mite Extract from Dermatophagoidesfarinae. J Invest Dermatol 2024; 144:1817-1828.e17. [PMID: 38302010 DOI: 10.1016/j.jid.2024.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 02/03/2024]
Abstract
Epidermal hyperinnervation is a critical feature of pruritus during skin inflammation. However, the mechanisms underlying epidermal hyperinnervation are unclear. This study investigates the role of the transcription factor EGR1 in epidermal innervation by utilizing wild-type (Egr1+/+) and Egr1-null (Egr1‒/‒) mice topically applied Dermatophagoides farinae extract from dust mite. Our findings revealed that Egr1‒/‒ mice exhibited reduced scratching behaviors and decreased density of epidermal innervation compared with Egr1+/+ mice. Furthermore, we identified artemin, a neurotrophic factor, as an EGR1 target responsible for Dermatophagoides farinae extract-induced hyperinnervation. It has been demonstrated that Dermatophagoides farinae extract stimulates toll-like receptors in keratinocytes. To elucidate the cellular mechanism, we stimulated keratinocytes with Pam3CSK4, a toll-like receptor 1/2 ligand. Pam3CSK4 triggered a toll-like receptor 1/2-mediated signaling cascade involving IRAK4, IκB kinase, MAPKs, ELK1, EGR1, and artemin, leading to increased neurite outgrowth and neuronal migration. In addition, increased expression of EGR1 and artemin was observed in the skin tissues of patients with atopic dermatitis. These findings highlight the significance of the EGR1-artemin axis in keratinocytes, promoting the process of epidermal innervation and suggesting it as a potential therapeutic target for alleviating itch and pain associated with house dust mite-induced skin inflammation.
Collapse
Affiliation(s)
- Hyunjin Yeo
- Department of Biological Sciences, Sang-huh College of Life Science, Konkuk University, Seoul, Republic of Korea
| | - Sung Shin Ahn
- Department of Biological Sciences, Sang-huh College of Life Science, Konkuk University, Seoul, Republic of Korea
| | - Sukjin Ou
- Department of Biological Sciences, Sang-huh College of Life Science, Konkuk University, Seoul, Republic of Korea
| | - Sook Jung Yun
- Department of Dermatology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yoongho Lim
- Division of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Dongsoo Koh
- Department of Applied Chemistry, Dongduk Women's University, Seoul, Republic of Korea
| | - Young Han Lee
- Department of Biological Sciences, Sang-huh College of Life Science, Konkuk University, Seoul, Republic of Korea; Cancer and Metabolism Institute, Konkuk University, Seoul, Republic of Korea
| | - Soon Young Shin
- Department of Biological Sciences, Sang-huh College of Life Science, Konkuk University, Seoul, Republic of Korea; Cancer and Metabolism Institute, Konkuk University, Seoul, Republic of Korea.
| |
Collapse
|
24
|
Li Y, Xu C, Qian X, Wang G, Han C, Hua H, Dong M, Chen J, Yu H, Zhang R, Feng X, Yang Z, Pan Y. Myeloid PTEN loss affects the therapeutic response by promoting stress granule assembly and impairing phagocytosis by macrophages in breast cancer. Cell Death Discov 2024; 10:344. [PMID: 39080255 PMCID: PMC11289284 DOI: 10.1038/s41420-024-02094-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024] Open
Abstract
Breast cancer (BRCA) has become the most common type of cancer in women. Improving the therapeutic response remains a challenge. Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) is a classic tumour suppressor with emerging new functions discovered in recent years, and myeloid PTEN loss has been reported to impair antitumour immunity. In this study, we revealed a novel mechanism by which myeloid PTEN potentially affects antitumour immunity in BRCA. We detected accelerated stress granule (SG) assembly under oxidative stress in PTEN-deficient bone marrow-derived macrophages (BMDMs) through the EGR1-promoted upregulation of TIAL1 transcription. PI3K/AKT/mTOR (PAM) pathway activation also promoted SG formation. ATP consumption during SG assembly in BMDMs impaired the phagocytic ability of 4T1 cells, potentially contributing to the disruption of antitumour immunity. In a BRCA neoadjuvant cohort, we observed a poorer response in myeloid PTENlow patients with G3BP1 aggregating as SGs in CD68+ cells, a finding that was consistent with the observation in our study that PTEN-deficient macrophages tended to more readily assemble SGs with impaired phagocytosis. Our results revealed the unconventional impact of SGs on BMDMs and might provide new perspectives on drug resistance and therapeutic strategies for the treatment of BRCA patients.
Collapse
Affiliation(s)
- Yan Li
- Department of Clinical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Chao Xu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Xiaojun Qian
- Department of Clinical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Gang Wang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Chaoqiang Han
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Hui Hua
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Menghao Dong
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Jian Chen
- Department of Clinical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Haiyang Yu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Rutong Zhang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Xiaoxi Feng
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Zhenye Yang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| | - Yueyin Pan
- Department of Clinical Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
25
|
Hyder U, Challa A, Thornton M, Nandu T, Kraus WL, D'Orso I. KAP1 negatively regulates RNA polymerase II elongation kinetics to activate signal-induced transcription. Nat Commun 2024; 15:5859. [PMID: 38997286 PMCID: PMC11245487 DOI: 10.1038/s41467-024-49905-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Signal-induced transcriptional programs regulate critical biological processes through the precise spatiotemporal activation of Immediate Early Genes (IEGs); however, the mechanisms of transcription induction remain poorly understood. By combining an acute depletion system with several genomics approaches to interrogate synchronized, temporal transcription, we reveal that KAP1/TRIM28 is a first responder that fulfills the temporal and heightened transcriptional demand of IEGs. Acute KAP1 loss triggers an increase in RNA polymerase II elongation kinetics during early stimulation time points. This elongation defect derails the normal progression through the transcriptional cycle during late stimulation time points, ultimately leading to decreased recruitment of the transcription apparatus for re-initiation thereby dampening IEGs transcriptional output. Collectively, KAP1 plays a counterintuitive role by negatively regulating transcription elongation to support full activation across multiple transcription cycles of genes critical for cell physiology and organismal functions.
Collapse
Affiliation(s)
- Usman Hyder
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ashwini Challa
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Micah Thornton
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tulip Nandu
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - W Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Iván D'Orso
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
26
|
Moghe M, Kim SS, Guan M, Rait A, Pirollo KF, Harford JB, Chang EH. scL-2PAM: A Novel Countermeasure That Ameliorates Neuroinflammation and Neuronal Losses in Mice Exposed to an Anticholinesterase Organophosphate. Int J Mol Sci 2024; 25:7539. [PMID: 39062781 PMCID: PMC11276659 DOI: 10.3390/ijms25147539] [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: 05/26/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Due to their inhibition of acetylcholinesterase, organophosphates are among the most toxic of chemicals. Pralidoxime (a.k.a 2-PAM) is the only acetylcholinesterase reactivator approved in the U.S., but 2-PAM only poorly traverses the blood-brain barrier. Previously, we have demonstrated that scL-2PAM, a nanoformulation designed to enter the brain via receptor-mediated transcytosis, is superior to unencapsulated 2-PAM for reactivating brain acetylcholinesterase, ameliorating cholinergic crisis, and improving survival rates for paraoxon-exposed mice. Here, we employ histology and transcriptome analyses to assess the ability of scL-2PAM to prevent neurological sequelae including microglial activation, expression of inflammatory cytokines, and ultimately loss of neurons in mice surviving paraoxon exposures. Levels of the mRNA encoding chemokine ligand 2 (CCL2) were significantly upregulated after paraoxon exposures, with CCL2 mRNA levels in the brain correlating well with the intensity and duration of cholinergic symptoms. Our nanoformulation of 2-PAM was found to be superior to unencapsulated 2-PAM in reducing the levels of the CCL2 transcript. Moreover, brain histology revealed that scL-2PAM was more effective than unencapsulated 2-PAM in preventing microglial activation and the subsequent loss of neurons. Thus, scL-2PAM appears to be a new and improved countermeasure for reducing neuroinflammation and mitigating brain damage in survivors of organophosphate exposures.
Collapse
Affiliation(s)
- Manish Moghe
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
| | - Sang-Soo Kim
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
- SynerGene Therapeutics, Inc., Potomac, MD 20854, USA;
| | - Miaoyin Guan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
| | - Antonina Rait
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
| | - Kathleen F. Pirollo
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
| | | | - Esther H. Chang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (M.M.); (A.R.); (K.F.P.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
27
|
Wang Y, Shen Z, Mo S, Zhang H, Chen J, Zhu C, Lv S, Zhang D, Huang X, Gu Y, Yu X, Ding X, Zhang X. Crosstalk among proximal tubular cells, macrophages, and fibroblasts in acute kidney injury: single-cell profiling from the perspective of ferroptosis. Hum Cell 2024; 37:1039-1055. [PMID: 38753279 PMCID: PMC11194220 DOI: 10.1007/s13577-024-01072-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/27/2024] [Indexed: 06/24/2024]
Abstract
The link between ferroptosis, a form of cell death mediated by iron and acute kidney injury (AKI) is recently gaining widespread attention. However, the mechanism of the crosstalk between cells in the pathogenesis and progression of acute kidney injury remains unexplored. In our research, we performed a non-negative matrix decomposition (NMF) algorithm on acute kidney injury single-cell RNA sequencing data based specifically focusing in ferroptosis-associated genes. Through a combination with pseudo-time analysis, cell-cell interaction analysis and SCENIC analysis, we discovered that proximal tubular cells, macrophages, and fibroblasts all showed associations with ferroptosis in different pathways and at various time. This involvement influenced cellular functions, enhancing cellular communication and activating multiple transcription factors. In addition, analyzing bulk expression profiles and marker genes of newly defined ferroptosis subtypes of cells, we have identified crucial cell subtypes, including Egr1 + PTC-C1, Jun + PTC-C3, Cxcl2 + Mac-C1 and Egr1 + Fib-C1. All these subtypes which were found in AKI mice kidneys and played significantly distinct roles from those of normal mice. Moreover, we verified the differential expression of Egr1, Jun, and Cxcl2 in the IRI mouse model and acute kidney injury human samples. Finally, our research presented a novel analysis of the crosstalk of proximal tubular cells, macrophages and fibroblasts in acute kidney injury targeting ferroptosis, therefore, contributing to better understanding the acute kidney injury pathogenesis, self-repairment and acute kidney injury-chronic kidney disease (AKI-CKD) progression.
Collapse
Affiliation(s)
- Yulin Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Medical Center of Kidney Disease, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Ziyan Shen
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Medical Center of Kidney Disease, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Institute of Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Shaocong Mo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Han Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Medical Center of Kidney Disease, No. 180 Fenglin Road, Shanghai, 200032, China
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jing Chen
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Medical Center of Kidney Disease, No. 180 Fenglin Road, Shanghai, 200032, China
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Cheng Zhu
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Medical Center of Kidney Disease, No. 180 Fenglin Road, Shanghai, 200032, China
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Shiqi Lv
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Medical Center of Kidney Disease, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Di Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Medical Center of Kidney Disease, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Xinhui Huang
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Medical Center of Kidney Disease, No. 180 Fenglin Road, Shanghai, 200032, China
- Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China
| | - Yulu Gu
- Division of Nephrology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213100, Jiangsu, China
| | - Xixi Yu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
- Shanghai Medical Center of Kidney Disease, No. 180 Fenglin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China.
- Shanghai Institute of Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China.
| | - Xiaoyan Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Shanghai, 200032, China.
- Shanghai Medical Center of Kidney Disease, No. 180 Fenglin Road, Shanghai, 200032, China.
- Shanghai Key Laboratory of Kidney and Blood Purification, No. 180 Fenglin Road, Shanghai, 200032, China.
- Shanghai Institute of Kidney and Dialysis, No. 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
28
|
Edalat SG, Gerber R, Houtman M, Lückgen J, Teixeira RL, Palacios Cisneros MDP, Pfanner T, Kuret T, Ižanc N, Micheroli R, Polido-Pereira J, Saraiva F, Lingam S, Burki K, Burja B, Pauli C, Rotar Ž, Tomšič M, Čučnik S, Fonseca JE, Distler O, Calado Â, Romão VC, Ospelt C, Sodin-Semrl S, Robinson MD, Frank Bertoncelj M. Molecular maps of synovial cells in inflammatory arthritis using an optimized synovial tissue dissociation protocol. iScience 2024; 27:109707. [PMID: 38832018 PMCID: PMC11144743 DOI: 10.1016/j.isci.2024.109707] [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: 07/21/2023] [Revised: 02/25/2024] [Accepted: 04/06/2024] [Indexed: 06/05/2024] Open
Abstract
In this study, we optimized the dissociation of synovial tissue biopsies for single-cell omics studies and created a single-cell atlas of human synovium in inflammatory arthritis. The optimized protocol allowed consistent isolation of highly viable cells from tiny fresh synovial biopsies, minimizing the synovial biopsy drop-out rate. The synovium scRNA-seq atlas contained over 100,000 unsorted synovial cells from 25 synovial tissues affected by inflammatory arthritis, including 16 structural, 11 lymphoid, and 15 myeloid cell clusters. This synovial cell map expanded the diversity of synovial cell types/states, detected synovial neutrophils, and broadened synovial endothelial cell classification. We revealed tissue-resident macrophage subsets with proposed matrix-sensing (FOLR2+COLEC12high) and iron-recycling (LYVE1+SLC40A1+) activities and identified fibroblast subsets with proposed functions in cartilage breakdown (SOD2highSAA1+SAA2+SDC4+) and extracellular matrix remodeling (SERPINE1+COL5A3+LOXL2+). Our study offers an efficient synovium dissociation method and a reference scRNA-seq resource, that advances the current understanding of synovial cell heterogeneity in inflammatory arthritis.
Collapse
Affiliation(s)
- Sam G. Edalat
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich and University of Zurich, 8952 Schlieren, Switzerland
| | - Reto Gerber
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich and University of Zurich, 8952 Schlieren, Switzerland
- Department of Molecular Life Sciences and SIB, Swiss Institute of Bioinformatics, University of Zurich, 8057 Zurich, Switzerland
| | - Miranda Houtman
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich and University of Zurich, 8952 Schlieren, Switzerland
| | | | - Rui Lourenço Teixeira
- Instituto de Medicina Molecular (iMM) João Lobo Antunes, Faculdade de Medicina, University of Lisbon, 1649-028 Lisbon, Portugal
- Faculdade de Medicina, University of Lisbon, 1649-028 Lisbon, Portugal
- Rheumatology Department, Hospital de Santa Maria, Lisbon Academic Medical Center, 1649-028 Lisbon, Portugal
| | | | | | - Tadeja Kuret
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich and University of Zurich, 8952 Schlieren, Switzerland
- Department of Rheumatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Nadja Ižanc
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich and University of Zurich, 8952 Schlieren, Switzerland
- Department of Rheumatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Raphael Micheroli
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich and University of Zurich, 8952 Schlieren, Switzerland
| | - Joaquim Polido-Pereira
- Instituto de Medicina Molecular (iMM) João Lobo Antunes, Faculdade de Medicina, University of Lisbon, 1649-028 Lisbon, Portugal
- Faculdade de Medicina, University of Lisbon, 1649-028 Lisbon, Portugal
- Rheumatology Department, Hospital de Santa Maria, Lisbon Academic Medical Center, 1649-028 Lisbon, Portugal
| | - Fernando Saraiva
- Instituto de Medicina Molecular (iMM) João Lobo Antunes, Faculdade de Medicina, University of Lisbon, 1649-028 Lisbon, Portugal
- Faculdade de Medicina, University of Lisbon, 1649-028 Lisbon, Portugal
- Rheumatology Department, Hospital de Santa Maria, Lisbon Academic Medical Center, 1649-028 Lisbon, Portugal
| | - Swathi Lingam
- Team PTA, BioMed X Institute, 69120 Heidelberg, Germany
| | - Kristina Burki
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich and University of Zurich, 8952 Schlieren, Switzerland
| | - Blaž Burja
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich and University of Zurich, 8952 Schlieren, Switzerland
- Department of Rheumatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Chantal Pauli
- Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Žiga Rotar
- Department of Rheumatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Matija Tomšič
- Department of Rheumatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Saša Čučnik
- Department of Rheumatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - João Eurico Fonseca
- Instituto de Medicina Molecular (iMM) João Lobo Antunes, Faculdade de Medicina, University of Lisbon, 1649-028 Lisbon, Portugal
- Faculdade de Medicina, University of Lisbon, 1649-028 Lisbon, Portugal
- Rheumatology Department, Hospital de Santa Maria, Lisbon Academic Medical Center, 1649-028 Lisbon, Portugal
| | - Oliver Distler
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich and University of Zurich, 8952 Schlieren, Switzerland
| | - Ângelo Calado
- Instituto de Medicina Molecular (iMM) João Lobo Antunes, Faculdade de Medicina, University of Lisbon, 1649-028 Lisbon, Portugal
| | - Vasco C. Romão
- Instituto de Medicina Molecular (iMM) João Lobo Antunes, Faculdade de Medicina, University of Lisbon, 1649-028 Lisbon, Portugal
- Faculdade de Medicina, University of Lisbon, 1649-028 Lisbon, Portugal
- Rheumatology Department, Hospital de Santa Maria, Lisbon Academic Medical Center, 1649-028 Lisbon, Portugal
| | - Caroline Ospelt
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich and University of Zurich, 8952 Schlieren, Switzerland
| | - Snežna Sodin-Semrl
- Department of Rheumatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, 6000 Koper, Slovenia
| | - Mark D. Robinson
- Department of Molecular Life Sciences and SIB, Swiss Institute of Bioinformatics, University of Zurich, 8057 Zurich, Switzerland
| | - Mojca Frank Bertoncelj
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich and University of Zurich, 8952 Schlieren, Switzerland
- Department of Molecular Life Sciences and SIB, Swiss Institute of Bioinformatics, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
29
|
Ferreté-Bonastre AG, Martínez-Gallo M, Morante-Palacios O, Calvillo CL, Calafell-Segura J, Rodríguez-Ubreva J, Esteller M, Cortés-Hernández J, Ballestar E. Disease activity drives divergent epigenetic and transcriptomic reprogramming of monocyte subpopulations in systemic lupus erythematosus. Ann Rheum Dis 2024; 83:865-878. [PMID: 38413168 DOI: 10.1136/ard-2023-225433] [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: 12/17/2023] [Accepted: 02/15/2024] [Indexed: 02/29/2024]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) is characterised by systemic inflammation involving various immune cell types. Monocytes, pivotal in promoting and regulating inflammation in SLE, differentiate from classic monocytes into intermediate and non-classic monocytes, assuming diverse roles and changing their proportions in inflammation. In this study, we investigated the epigenetic and transcriptomic profiles of these and novel monocyte subsets in SLE in relation to activity and progression. METHODS We obtained the DNA methylomes and transcriptomes of classic, intermediate, non-classic monocytes in patients with SLE (at first and follow-up visits) and healthy donors. We integrated these data with single-cell transcriptomics of SLE and healthy donors and interrogated their relationships with activity and progression. RESULTS In addition to shared DNA methylation and transcriptomic alterations associated with a strong interferon signature, we identified monocyte subset-specific alterations, especially in DNA methylation, which reflect an impact of SLE on monocyte differentiation. SLE classic monocytes exhibited a proinflammatory profile and were primed for macrophage differentiation. SLE non-classic monocytes displayed a T cell differentiation-related phenotype, with Th17-regulating features. Changes in monocyte proportions, DNA methylation and expression occurred in relation to disease activity and involved the STAT pathway. Integration of bulk with single-cell RNA sequencing datasets revealed disease activity-dependent expansion of SLE-specific monocyte subsets, further supported the interferon signature for classic monocytes, and associated intermediate and non-classic populations with exacerbated complement activation. CONCLUSIONS Disease activity in SLE drives a subversion of the epigenome and transcriptome programme in monocyte differentiation, impacting the function of different subsets and allowing to generate predictive methods for activity and progression.
Collapse
Affiliation(s)
| | - Mónica Martínez-Gallo
- Immunology Division, Vall d'Hebron University Hospital and Diagnostic Immunology Research Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | | | - Celia Lourdes Calvillo
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Josep Calafell-Segura
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Javier Rodríguez-Ubreva
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Cancer (CIBERONC), Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Spain
| | - Josefina Cortés-Hernández
- Rheumatology Department, Hospital Vall d'Hebron and Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
- Epigenetics in Inflammatory and Metabolic Diseases Laboratory, Health Science Center (HSC), East China Normal University (ECNU), Shanghai, China
| |
Collapse
|
30
|
Hyder U, Challa A, Thornton M, Nandu T, Kraus WL, D’Orso I. KAP1 negatively regulates RNA polymerase II elongation kinetics to activate signal-induced transcription. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.05.592422. [PMID: 38746145 PMCID: PMC11092767 DOI: 10.1101/2024.05.05.592422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Signal-induced transcriptional programs regulate critical biological processes through the precise spatiotemporal activation of Immediate Early Genes (IEGs); however, the mechanisms of transcription induction remain poorly understood. By combining an acute depletion system with high resolution genomics approaches to interrogate synchronized, temporal transcription, we reveal that KAP1/TRIM28 is a first responder that fulfills the temporal and heightened transcriptional demand of IEGs. Unexpectedly, acute KAP1 loss triggers an increase in RNA polymerase II elongation kinetics during early stimulation time points. This elongation defect derails the normal progression through the transcriptional cycle during late stimulation time points, ultimately leading to decreased recruitment of the transcription apparatus for re-initiation thereby dampening IEGs transcriptional output. Collectively, KAP1 plays a counterintuitive role by negatively regulating transcription elongation to support full activation across multiple transcription cycles of genes critical for cell physiology and organismal functions.
Collapse
Affiliation(s)
- Usman Hyder
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ashwini Challa
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Micah Thornton
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tulip Nandu
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - W. Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Iván D’Orso
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
31
|
Hashoul D, Saliba W, Broza YY, Haick H. Non-contact immunological signaling for highly-efficient regulation of the transcriptional map of human monocytes. Bioeng Transl Med 2024; 9:e10519. [PMID: 38818125 PMCID: PMC11135151 DOI: 10.1002/btm2.10519] [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: 01/05/2023] [Revised: 03/18/2023] [Accepted: 03/29/2023] [Indexed: 06/01/2024] Open
Abstract
The different immune system cells communicate and coordinate a response using a complex and evolved language of cytokines and chemokines. These cellular interactions carry out multiple functions in distinct cell types with numerous developmental outcomes. Despite the plethora of different cytokines and their cognate receptors, there is a restricted number of signal transducers and activators to control immune responses. Herein, we report on a new class of immunomodulatory signaling molecules based on volatile molecules (VMs, namely, volatile organic compounds [VOCs]), by which they can affect and/or control immune cell behavior and transcriptomic profile without any physical contact with other cells. The study demonstrates the role of VMs by analyzing non-contact cell communication between normal and cancerous lung cells and U937 monocytes, which are key players in the tumor microenvironment. Integrated transcriptome and proteome analyses showed the suggested regulatory role of VMs released from normal and cancer cells on neighboring monocytes in several molecular pathways, including PI3K/AKT, PPAR, and HIF-1. Presented data provide an initial platform for a new class of immunomodulatory molecules that can potentially mirror the genomic and proteomic profile of cells, thereby paving the way toward non-invasive immunomonitoring.
Collapse
Affiliation(s)
- Dina Hashoul
- Department of Chemical Engineering and Russell Berrie Nanotechnology InstituteTechnion ‐ Israel Institute of TechnologyHaifaIsrael
| | - Walaa Saliba
- Department of Chemical Engineering and Russell Berrie Nanotechnology InstituteTechnion ‐ Israel Institute of TechnologyHaifaIsrael
| | - Yoav Y. Broza
- Department of Chemical Engineering and Russell Berrie Nanotechnology InstituteTechnion ‐ Israel Institute of TechnologyHaifaIsrael
| | - Hossam Haick
- Department of Chemical Engineering and Russell Berrie Nanotechnology InstituteTechnion ‐ Israel Institute of TechnologyHaifaIsrael
| |
Collapse
|
32
|
Jiang S, Lu H, Pan Y, Yang A, Aikemu A, Li H, Hao R, Huang Q, Qi X, Tao Z, Wu Y, Quan C, Zhou G, Lu Y. Characterization of the distinct immune microenvironments between hepatocellular carcinoma and intrahepatic cholangiocarcinoma. Cancer Lett 2024; 588:216799. [PMID: 38479553 DOI: 10.1016/j.canlet.2024.216799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/06/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
As two major types of primary liver cancers, the tumor immune microenvironment (TIME) of hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC) have been well studied separately. However, a systemic assessment of the similarities and differences between the TIME of HCC and ICC is still lacking. In this study, we pictured a landscape of combined TIME of HCC and ICC by sequencing and integrating 41 single-cell RNA-seq samples from four different tissue types of both malignancies. We found that T cells in HCC tumors generally exhibit higher levels of immunosuppression and exhaustion than those in ICC tumors. Myeloid cells in HCC and ICC tumors also exhibit distinct phenotypes and may serve as a key factor driving the differences between their TIMEs. Besides, we identified a cluster of EGR1+ macrophages specifically enriched in HCC tumors. Together, our study provides new insights into cellular composition, states and interactions in the TIMEs of HCC and ICC, which could pave the way for the development of future therapeutic targets for liver cancers.
Collapse
Affiliation(s)
- Siao Jiang
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China; School of Life Science, University of Hebei, Baoding City, Hebei Province, PR China
| | - Hao Lu
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Yingwei Pan
- Department of Hepatobiliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, PR China
| | - Aiqing Yang
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Ainiwaer Aikemu
- College of Xinjiang Uyghur Medicine, Hetian City, Xinjiang Province, PR China
| | - Hao Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Rongjiao Hao
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China; School of Life Science, University of Hebei, Baoding City, Hebei Province, PR China
| | - Qilin Huang
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China; Collaborative Innovation Center for Personalized Cancer Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing City, Jiangsu Province, PR China
| | - Xin Qi
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China; Medical College, Guizhou University, Guiyang City, Guizhou Province, PR China
| | - Zongjian Tao
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Yinglong Wu
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Cheng Quan
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China.
| | - Gangqiao Zhou
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China; School of Life Science, University of Hebei, Baoding City, Hebei Province, PR China; Collaborative Innovation Center for Personalized Cancer Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing City, Jiangsu Province, PR China; Medical College, Guizhou University, Guiyang City, Guizhou Province, PR China.
| | - Yiming Lu
- State Key Laboratory of Proteomics, National Center for Protein Sciences at Beijing, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, PR China; School of Life Science, University of Hebei, Baoding City, Hebei Province, PR China.
| |
Collapse
|
33
|
Challagundla N, Shah D, Dalai SK, Agrawal-Rajput R. IFNγ insufficiency during mouse intra-vaginal Chlamydia trachomatis infection exacerbates alternative activation in macrophages with compromised CD40 functions. Int Immunopharmacol 2024; 131:111821. [PMID: 38484664 DOI: 10.1016/j.intimp.2024.111821] [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: 11/04/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/10/2024]
Abstract
Chlamydia trachomatis (C.tr), an obligate intracellular pathogen, causes asymptomatic genital infections in women and is a leading cause of preventable blindness. We have developed in vivo mouse models of acute and chronic C. trachomatis genital infection to explore the significance of macrophage-directed response in mediating immune activation/suppression. Our findings reveal that during chronic and repeated C. trachomatis infections, Th1 response is abated while Treg response is enhanced. Additionally, an increase in exhaustion (PD1, CTLA4) and anergic (Klrg3, Tim3) T cell markers is observed during chronic infection. We have also observed that M2 macrophages with low CD40 expression promote Th2 and Treg differentiation leading to sustained C. trachomatis genital infection. Macrophages infected with C. trachomatis or treated with supernatant of infected epithelial cells drive them to an M2 phenotype. C. trachomatis infection prevents the increase in CD40 expression as observed in western blots and flow cytometric analysis. Insufficient IFNγ, as observed during chronic infection, leads to incomplete clearance of bacteria and poor immune activation. C. trachomatis decapacitates IFNγ responsiveness in macrophages via hampering IFNγRI and IFNγRII expression which can be correlated with poor expression of MHC-II, CD40, iNOS and NO release even following IFNγ supplementation. M2 macrophages during C. trachomatis infection express low CD40 rendering immunosuppressive, Th2 and Treg differentiation which could not be reverted even by IFNγ supplementation. The alternative macrophages also harbour high bacterial load and are poor responders to IFNγ, thus promoting immunosuppression. In summary, C. trachomatis modulates the innate immune cells, attenuating the anti-chlamydial functions of T cells in a manner that involves decreased CD40 expression on macrophages.
Collapse
Affiliation(s)
- Naveen Challagundla
- Immunology Lab, Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India.
| | - Dhruvi Shah
- Immunology Lab, Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India.
| | - Sarat K Dalai
- Institute of Science, Nirma University, S.G. Highway, Ahmedabad, Gujarat, India.
| | - Reena Agrawal-Rajput
- Immunology Lab, Biological Sciences and Biotechnology, Indian Institute of Advanced Research, Gandhinagar, Gujarat, India.
| |
Collapse
|
34
|
Wang H, Tian Q, Zhang R, Du Q, Hu J, Gao T, Gao S, Fan K, Cheng X, Yan S, Zheng G, Dong H. Nobiletin alleviates atherosclerosis by inhibiting lipid uptake via the PPARG/CD36 pathway. Lipids Health Dis 2024; 23:76. [PMID: 38468335 PMCID: PMC10926578 DOI: 10.1186/s12944-024-02049-5] [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: 12/01/2023] [Accepted: 02/18/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Atherosclerosis (AS) is a persistent inflammatory condition triggered and exacerbated by several factors including lipid accumulation, endothelial dysfunction and macrophages infiltration. Nobiletin (NOB) has been reported to alleviate atherosclerosis; however, the underlying mechanism remains incompletely understood. METHODS This study involved comprehensive bioinformatic analysis, including multidatabase target prediction; GO and KEGG enrichment analyses for function and pathway exploration; DeepSite and AutoDock for drug binding site prediction; and CIBERSORT for immune cell involvement. In addition, target intervention was verified via cell scratch assays, oil red O staining, ELISA, flow cytometry, qRT‒PCR and Western blotting. In addition, by establishing a mouse model of AS, it was demonstrated that NOB attenuated lipid accumulation and the extent of atherosclerotic lesions. RESULTS (1) Altogether, 141 potentially targetable genes were identified through which NOB could intervene in atherosclerosis. (2) Lipid and atherosclerosis, fluid shear stress and atherosclerosis may be the dominant pathways and potential mechanisms. (3) ALB, AKT1, CASP3 and 7 other genes were identified as the top 10 target genes. (4) Six genes, including PPARG, MMP9, SRC and 3 other genes, were related to the M0 fraction. (5) CD36 and PPARG were upregulated in atherosclerosis samples compared to the normal control. (6) By inhibiting lipid uptake in RAW264.7 cells, NOB prevents the formation of foam cell. (7) In RAW264.7 cells, the inhibitory effect of oxidized low-density lipoprotein on foam cells formation and lipid accumulation was closely associated with the PPARG signaling pathway. (8) In vivo validation showed that NOB significantly attenuated intra-arterial lipid accumulation and macrophage infiltration and reduced CD36 expression. CONCLUSIONS Nobiletin alleviates atherosclerosis by inhibiting lipid uptake via the PPARG/CD36 pathway.
Collapse
Affiliation(s)
- Heng Wang
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qinqin Tian
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruijing Zhang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qiujing Du
- Jiangyin People's Hospital, Wuxi, Jiangsu, China
- Shanxi Bethune Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Jie Hu
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Tingting Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Siqi Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Keyi Fan
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xing Cheng
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Sheng Yan
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guoping Zheng
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.
| | - Honglin Dong
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
35
|
Wang W, Jiang J, Yang C, Meng X, Gao L, Yuan Y, Lei T, Ding P, Yin R, Li Q. A critical time window for leukapheresis product transportation to manufacture clinical-grade dendritic cells with optimal anti-tumor activities. Cytotherapy 2024; 26:210-220. [PMID: 38127032 DOI: 10.1016/j.jcyt.2023.12.003] [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: 05/15/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND AIMS Dendritic cell (DC)-based immunotherapy is a promising approach to treat cancer. However, key aspects governing the reproducible manufacturing of high-quality DC remain incompletely defined. Here, we show that the time window between leukapheresis and DC manufacturing is critical. METHODS Transcriptomic profiling by RNA-seq was used to unbiasedly characterize cellular states during each step of DC manufacturing process, and functional assays were used to determine the anti-tumor activities of DC. RESULTS During preclinical development of a DC-based cytotherapy platform, CUD-002 (NCT05270720), we found that DC quality varied among different batches, even though commonly used DC maturation markers CD80, CD83 and CD86 were indistinguishable. Multivariate analysis indicated that DC quality was negatively associated with the shipping time from the leukapheresis site to the manufacturing center. To investigate the potential effect of shipping time, we stored leukapheresis materials from three donors for 0, 1, 2 or 3 days before DC manufacturing. For each step, we carried out RNA-seq analysis to unbiasedly characterize cellular states. Integrated bioinformatic analyses indicated that longer storage time reduced the expression of several transcription factors to attenuate interferon pathways. CONCLUSIONS Consistently, we found that 3-day storage of leukapheresis materials significantly lowered the efficiency to generate DC but also impaired DC responses to inflammatory signals, resulting in inferior antigen-presentation and cytotoxic T-cell activities. Thus, we recommend using leukapheresis materials within 48 h to manufacture therapeutic DCs.
Collapse
Affiliation(s)
- Wenxiang Wang
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China; Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, China
| | - Jinfeng Jiang
- Non-coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, Sichuan, China
| | - Chao Yang
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Xiangjun Meng
- Division of Cell Manufacturing, Sichuan Cunde Therapeutics, Chengdu, Sichuan, China
| | - Li Gao
- Division of Cell Manufacturing, Sichuan Cunde Therapeutics, Chengdu, Sichuan, China
| | - Yuan Yuan
- Division of Cell Manufacturing, Sichuan Cunde Therapeutics, Chengdu, Sichuan, China
| | - Tingjun Lei
- Division of Cell Manufacturing, Sichuan Cunde Therapeutics, Chengdu, Sichuan, China
| | - Ping Ding
- Non-coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, Sichuan, China.
| | - Rutie Yin
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China.
| | - Qintong Li
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
36
|
Yang R, Wang X, Liu H, Chen J, Tan C, Chen H, Wang X. Egr-1 is a key regulator of the blood-brain barrier damage induced by meningitic Escherichia coli. Cell Commun Signal 2024; 22:44. [PMID: 38233877 PMCID: PMC10795328 DOI: 10.1186/s12964-024-01488-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/11/2024] [Indexed: 01/19/2024] Open
Abstract
Bacterial meningitis remains a leading cause of infection-related mortality worldwide. Although Escherichia coli (E. coli) is the most common etiology of neonatal meningitis, the underlying mechanisms governing bacterial blood-brain barrier (BBB) disruption during infection remain elusive. We observed that infection of human brain microvascular endothelial cells with meningitic E. coli triggers the activation of early growth response 1 (Egr-1), a host transcriptional activator. Through integrated chromatin immunoprecipitation sequencing and transcriptome analysis, we identified Egr-1 as a crucial regulator for maintaining BBB integrity. Mechanistically, Egr-1 induced cytoskeletal changes and downregulated tight junction protein expression by directly targeting VEGFA, PDGFB, and ANGPTL4, resulting in increased BBB permeability. Meanwhile, Egr-1 also served as a master regulator in the initiation of neuroinflammatory response during meningitic E. coli infection. Our findings support an Egr-1-dependent mechanism of BBB disruption by meningitic E. coli, highlighting a promising therapeutic target for bacterial meningitis.
Collapse
Affiliation(s)
- Ruicheng Yang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Xinyi Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Hulin Liu
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Jiaqi Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Chen Tan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, 430070, China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, 430070, China
| | - Xiangru Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, 430070, China.
- International Research Center for Animal Disease, Ministry of Science and Technology of the People's Republic of China, Wuhan, 430070, China.
| |
Collapse
|
37
|
Rocks D, Purisic E, Gallo EF, Greally JM, Suzuki M, Kundakovic M. Egr1 is a sex-specific regulator of neuronal chromatin, synaptic plasticity, and behaviour. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572697. [PMID: 38187614 PMCID: PMC10769422 DOI: 10.1101/2023.12.20.572697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Sex differences are found in brain structure and function across species, and across brain disorders in humans1-3. The major source of brain sex differences is differential secretion of steroid hormones from the gonads across the lifespan4. Specifically, ovarian hormones oestrogens and progesterone are known to dynamically change structure and function of the adult female brain, having a major impact on psychiatric risk5-7. However, due to limited molecular studies in female rodents8, very little is still known about molecular drivers of female-specific brain and behavioural plasticity. Here we show that overexpressing Egr1, a candidate oestrous cycle-dependent transcription factor9, induces sex-specific changes in ventral hippocampal neuronal chromatin, gene expression, and synaptic plasticity, along with hippocampus-dependent behaviours. Importantly, Egr1 overexpression mimics the high-oestrogenic phase of the oestrous cycle, and affects behaviours in ovarian hormone-depleted females but not in males. We demonstrate that Egr1 opens neuronal chromatin directly across the sexes, although with limited genomic overlap. Our study not only reveals the first sex-specific chromatin regulator in the brain, but also provides functional evidence that this sex-specific gene regulation drives neuronal gene expression, synaptic plasticity, and anxiety- and depression-related behaviour. Our study exemplifies an innovative sex-based approach to studying neuronal gene regulation1 in order to understand sex-specific synaptic and behavioural plasticity and inform novel brain disease treatments.
Collapse
Affiliation(s)
- Devin Rocks
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Eric Purisic
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Eduardo F. Gallo
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - John M. Greally
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Masako Suzuki
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Marija Kundakovic
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| |
Collapse
|
38
|
Leary JR, Bacher R. Interpretable trajectory inference with single-cell Linear Adaptive Negative-binomial Expression (scLANE) testing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.19.572477. [PMID: 38187622 PMCID: PMC10769309 DOI: 10.1101/2023.12.19.572477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The rapid proliferation of trajectory inference methods for single-cell RNA-seq data has allowed researchers to investigate complex biological processes by examining underlying gene expression dynamics. After estimating a latent cell ordering, statistical models are used to determine which genes exhibit changes in expression that are significantly associated with progression through the biological trajectory. While a few techniques for performing trajectory differential expression exist, most rely on the flexibility of generalized additive models in order to account for the inherent nonlinearity of changes in gene expression. As such, the results can be difficult to interpret, and biological conclusions often rest on subjective visual inspections of the most dynamic genes. To address this challenge, we propose scLANE testing, which is built around an interpretable generalized linear model and handles nonlinearity with basis splines chosen empirically for each gene. In addition, extensions to estimating equations and mixed models allow for reliable trajectory testing under complex experimental designs. After validating the accuracy of scLANE under several different simulation scenarios, we apply it to a set of diverse biological datasets and display its ability to provide novel biological information when used downstream of both pseudotime and RNA velocity estimation methods.
Collapse
Affiliation(s)
- Jack R. Leary
- Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, USA
| | - Rhonda Bacher
- Department of Biostatistics, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
39
|
Cantu A, Gutierrez MC, Dong X, Leek C, Anguera M, Lingappan K. Modulation of recovery from neonatal hyperoxic lung injury by sex as a biological variable. Redox Biol 2023; 68:102933. [PMID: 38661305 PMCID: PMC10628633 DOI: 10.1016/j.redox.2023.102933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 04/26/2024] Open
Abstract
Recovery from lung injury during the neonatal period requires the orchestration of many biological pathways. The modulation of such pathways can drive the developing lung towards proper repair or persistent maldevelopment that can lead to a disease phenotype. Sex as a biological variable can regulate these pathways differently in the male and female lung exposed to neonatal hyperoxia. In this study, we assessed the contribution of cellular diversity in the male and female neonatal lung following injury. Our objective was to investigate sex and cell-type specific transcriptional changes that drive repair or persistent injury in the neonatal lung and delineate the alterations in the immune-endothelial cell communication networks using single cell RNA sequencing (sc-RNAseq) in a murine model of hyperoxic injury. We generated transcriptional profiles of >55,000 cells isolated from the lungs of postnatal day 1 (PND 1; pre-exposure), PND 7, and PND 21neonatal male and female C57BL/6 mice exposed to 95 % FiO2 between PND 1-5 (saccular stage of lung development). We show the presence of sex-based differences in the transcriptional states of lung endothelial and immune cells at PND 1 and PND 21. Furthermore, we demonstrate that biological sex significantly influences the response to injury, with a greater number of differentially expressed genes showing sex-specific patterns than those shared between male and female lungs. Pseudotime trajectory analysis highlighted genes needed for lung development that were altered by hyperoxia. Finally, we show intercellular communication between endothelial and immune cells at saccular and alveolar stages of lung development with sex-based biases in the crosstalk and identify novel ligand-receptor pairs. Our findings provide valuable insights into the cell diversity, transcriptional state, developmental trajectory, and cell-cell communication underlying neonatal lung injury, with implications for understanding lung development and possible therapeutic interventions while highlighting the crucial role of sex as a biological variable.
Collapse
Affiliation(s)
- Abiud Cantu
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Xiaoyu Dong
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Connor Leek
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Montserrat Anguera
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Krithika Lingappan
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
40
|
Zhang M, Lv L, Luo H, Cai H, Yu L, Jiang Y, Gao F, Tong W, Li L, Li G, Zhou Y, Tong G, Liu C. The CD2v protein of African swine fever virus inhibits macrophage migration and inflammatory cytokines expression by downregulating EGR1 expression through dampening ERK1/2 activity. Vet Res 2023; 54:106. [PMID: 37968713 PMCID: PMC10648359 DOI: 10.1186/s13567-023-01239-w] [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: 07/11/2023] [Accepted: 10/12/2023] [Indexed: 11/17/2023] Open
Abstract
African swine fever virus (ASFV) is a highly contagious and deadly virus that leads to high mortality rates in domestic swine populations. Although the envelope protein CD2v of ASFV has been implicated in immunomodulation, the molecular mechanisms underlying CD2v-mediated immunoregulation remain unclear. In this study, we generated a stable CD2v-expressing porcine macrophage (PAM-CD2v) line and investigated the CD2v-dependent transcriptomic landscape using RNA-seq. GO terms enrichment analysis and gene set enrichment analysis revealed that CD2v predominantly affected the organization and assembly process of the extracellular matrix. Wound healing and Transwell assays showed that CD2v inhibited swine macrophage migration. Further investigation revealed a significant decrease in the expression of transcription factor early growth response 1 (EGR1) through inhibiting the activity of extracellular signal-regulated kinase 1 and 2 (ERK1/2). Notably, EGR1 knockout in swine macrophages restricted cell migration, whereas EGR1 overexpression in PAM-CD2v restored the ability of macrophage migration, suggesting that CD2v inhibits swine macrophage motility by downregulating EGR1 expression. Furthermore, we performed chromatin immunoprecipitation and sequencing for EGR1 and the histone mark H3K27 acetylation (H3K27ac), and we found that EGR1 co-localized with the activated histone modification H3K27ac neighboring the transcriptional start sites. Further analysis indicated that EGR1 and H3K27ac co-occupy the promoter regions of cell locomotion-related genes. Finally, by treating various derivatives of swine macrophages with lipopolysaccharides, we showed that depletion of EGR1 decreased the expression of inflammatory cytokines including TNFα, IL1α, IL1β, IL6, and IL8, which play essential roles in inflammation and host immune response. Collectively, our results provide new insights into the immunomodulatory mechanism of ASFV CD2v.
Collapse
Affiliation(s)
- Min Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Lilei Lv
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Huaye Luo
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
| | - Hongming Cai
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Lingxue Yu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Yifeng Jiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Fei Gao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Wu Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Liwei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Guoxin Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Yanjun Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Guangzhi Tong
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China.
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, 225009, China.
| | - Changlong Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, 200241, China.
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
41
|
Maria NI, Papoin J, Raparia C, Sun Z, Josselsohn R, Lu A, Katerji H, Syeda MM, Polsky D, Paulson R, Kalfa T, Barnes BJ, Zhang W, Blanc L, Davidson A. Human TLR8 induces inflammatory bone marrow erythromyeloblastic islands and anemia in SLE-prone mice. Life Sci Alliance 2023; 6:e202302241. [PMID: 37495396 PMCID: PMC10372407 DOI: 10.26508/lsa.202302241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/28/2023] Open
Abstract
Anemia commonly occurs in systemic lupus erythematosus, a disease characterized by innate immune activation by nucleic acids. Overactivation of cytoplasmic sensors by self-DNA or RNA can cause erythroid cell death, while sparing other hematopoietic cell lineages. Whereas chronic inflammation is involved in this mechanism, less is known about the impact of systemic lupus erythematosus on the BM erythropoietic niche. We discovered that expression of the endosomal ssRNA sensor human TLR8 induces fatal anemia in Sle1.Yaa lupus mice. We observed that anemia was associated with a decrease in erythromyeloblastic islands and a block in differentiation at the CFU-E to proerythroblast transition in the BM. Single-cell RNAseq analyses of isolated BM erythromyeloblastic islands from human TLR8-expressing mice revealed that genes associated with essential central macrophage functions including adhesion and provision of nutrients were down-regulated. Although compensatory stress erythropoiesis occurred in the spleen, red blood cell half-life decreased because of hemophagocytosis. These data implicate the endosomal RNA sensor TLR8 as an additional innate receptor whose overactivation causes acquired failure of erythropoiesis via myeloid cell dysregulation.
Collapse
Affiliation(s)
- Naomi I Maria
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| | - Julien Papoin
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| | - Chirag Raparia
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| | - Zeguo Sun
- Department of Medicine, Mount Sinai Medical Center, New York, NY, USA
| | - Rachel Josselsohn
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Ailing Lu
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hani Katerji
- Department of Pathology, University of Rochester, Rochester, NY, USA
| | - Mahrukh M Syeda
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, USA
| | - David Polsky
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, USA
| | - Robert Paulson
- Department of Veterinary and Biomedical Sciences, Penn State College of Agricultural Sciences, University Park, PA, USA
| | - Theodosia Kalfa
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Betsy J Barnes
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| | - Weijia Zhang
- Department of Medicine, Mount Sinai Medical Center, New York, NY, USA
| | - Lionel Blanc
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| | - Anne Davidson
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| |
Collapse
|
42
|
Shin S, Choi EJ, Moon SW, Lee SB, Chung YJ, Lee SH. Leprosy-specific subsets of macrophages and Schwann cells identified by single-cell RNA-sequencing. Pathol Res Pract 2023; 250:154821. [PMID: 37757621 DOI: 10.1016/j.prp.2023.154821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023]
Abstract
In Mycobacterium leprae (M. leprae)-infection, inflammatory cells' subsets and dynamics as well as the interactions with Schwann cells have remained elusive. We investigated individual cells in M. leprae-inoculated nude mice by single-cell RNA-sequencing (scRNA-seq). For macrophages, we dissected two M1-like subsets and five M2-like subsets, where lipid-associated signatures were pervasive in both M1-like and M2-like subsets. There were four macrophage trajectories showing: (i) pro-inflammatory (M1), (ii) lipid metabolism-related (M2), (iii) anti-inflammatory (M2), and (iv) interferon-stimulated gene-related (M2) fates. They displayed early divergence without ever rejoining along the paths, suggesting simultaneous or continuous stimuli for macrophage activation in leprosy. The scRNA-seq predicted Schwann cell-macrophage interactions (Notch1-Jag1, Plxnb1-Sema4d interactions). An immature Schwann cell subset showing Tfap2a expression was identified, indicating Schwann cell dedifferentiation in leprosy tissues. Expressions of Notch1, Jag1, Plxnb1, Sema4d, and Tfap2a were validated in mouse or human leprosy tissues by immunohistochemistry. We identified both pro-inflammatory and inflammation-resolution signatures, where lipid-associated signatures were pervasive to the macrophages, representing leprosy-specific macrophage states for prolonged and repeated episodes of inflammation and resolution. Our study identified refined molecular states and interactions of macrophages and Schwann cells, suggesting novel insights into the pathogenesis of unhealed inflammation with neuropathy and potential therapeutic targets for leprosy.
Collapse
Affiliation(s)
- Sun Shin
- Departments of Microbiology, College of Medicine, The Catholic University of Korea, Republic of Korea; Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Eun Ji Choi
- Departments of Pathology, College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Seong Won Moon
- Departments of Pathology, College of Medicine, The Catholic University of Korea, Republic of Korea; Departments of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Seong-Beom Lee
- Institute of Hansen's Disease, College of Medicine, The Catholic University of Korea, Republic of Korea; Departments of Pathology, College of Medicine, The Catholic University of Korea, Republic of Korea
| | - Yeun-Jun Chung
- Departments of Microbiology, College of Medicine, The Catholic University of Korea, Republic of Korea; Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Republic of Korea; Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Republic of Korea.
| | - Sug Hyung Lee
- Departments of Pathology, College of Medicine, The Catholic University of Korea, Republic of Korea; Departments of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Republic of Korea; Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Republic of Korea.
| |
Collapse
|
43
|
Wang C, Zhang X, Chen R, Zhu X, Lian N. EGR1 mediates METTL3/m 6A/CHI3L1 to promote osteoclastogenesis in osteoporosis. Genomics 2023; 115:110696. [PMID: 37558013 DOI: 10.1016/j.ygeno.2023.110696] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 07/27/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023]
Abstract
OBJECTIVE To investigate EGR1-mediated METTL3/m6A/CHI3L1 axis in osteoporosis. METHODS Ovariectomy (OVX) was performed on mice to induce osteoporosis, followed by μ-CT scanning of femurs, histological staining, immunohistochemistry analysis of MMP9 and NFATc1, and ELISA of serum BGP, ALP, Ca, and CTXI. The isolated mouse bone marrow mononuclear macrophages (BMMs) were differentiated into osteoclasts under cytokine stimulation. TRAP staining was performed to quantify osteoclasts. The levels of Nfatc1, c-Fos, Acp5, and Ctsk in osteoclasts, m6A level, and the relationships among EGR1, METTL3, and CHI3L1 were analyzed. RESULTS The EGR1/METTL3/CHI3L1 levels and m6A level were upregulated in osteoporotic mice and the derived BMMs. EGR1 was a transcription factor of METTL3. METTL3 promoted the post-transcriptional regulation of CHI3L1 by increasing m6A methylation. EGR1 downregulation reduced BMMs-differentiated osteoclasts and alleviated OVX-induced osteoporosis by regulating the METTL3/m6A/CHI3L1 axis. CONCLUSION EGR1 promotes METTL3 transcription and increases m6A-modified CHI3L1 level, thereby stimulating osteoclast differentiation and osteoporosis development.
Collapse
Affiliation(s)
- Changsheng Wang
- Department of Spinal Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, PR China.
| | - Xiaobo Zhang
- Department of Spinal Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, PR China
| | - Rongsheng Chen
- Department of Spinal Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, PR China
| | - Xitian Zhu
- Department of Spinal Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, PR China
| | - Nancheng Lian
- Department of Spinal Surgery, First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350005, PR China
| |
Collapse
|
44
|
Shen L, Yin H, Sun L, Zhang Z, Jin Y, Cao S, Fu Q, Fan C, Bao C, Lu L, Zhan Y, Xu X, Chen X, Yan Q. Iguratimod attenuated fibrosis in systemic sclerosis via targeting early growth response 1 expression. Arthritis Res Ther 2023; 25:151. [PMID: 37596660 PMCID: PMC10439582 DOI: 10.1186/s13075-023-03135-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 08/02/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND The early growth response 1 (EGR1) is a central transcription factor involved in systemic sclerosis (SSc) pathogenesis. Iguratimod is a synthesized anti-rheumatic disease-modifying drug, which shows drastic inhibition to EGR1 expression in B cells. This study is aiming to investigate the anti-fibrotic effect of iguratimod in SSc. METHODS EGR1 was detected by immunofluorescence staining real-time PCR or western blot. Iguratimod was applied in EGR1 overexpressed or knockdown human dermal fibroblast, bleomycin pre-treated mice, tight skin 1 mice, and SSc skin xenografts. RNA sequencing was performed in cultured fibroblast and xenografts to identify the iguratimod regulated genes. RESULTS EGR1 overexpressed predominantly in non-immune cells of SSc patients. Iguratimod reduced EGR1 expression in fibroblasts and neutralized changes of EGR1 response genes regulated by TGFβ. The extracellular matrix (ECM) production and activation of fibroblasts were attenuated by iguratimod while EGR1 overexpression reversed this effect of iguratimod. Iguratimod ameliorated the skin fibrosis induced by bleomycin and hypodermal fibrosis in TSK-1 mice. Decreasing in the collagen content as well as the density of EGR1 or TGFβ positive fibroblasts of skin xenografts from naïve SSc patients was observed after local treatment of iguratimod. CONCLUSION Targeting EGR1 expression is a probable underlying mechanism for the anti-fibrotic effect of iguratimod.
Collapse
Affiliation(s)
- Lichong Shen
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Hanlin Yin
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Li Sun
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhiliang Zhang
- Department of Plastic Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Yuyang Jin
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Shan Cao
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Qiong Fu
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Chaofan Fan
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Chunde Bao
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Liangjing Lu
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Yifan Zhan
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, 201203, China
| | - Xiaojiang Xu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA.
| | - Xiaoxiang Chen
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China.
- Department of Rheumatology, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong Universuty, Nantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong Universuty School of Medicine, Nantong, 226006, China.
| | - Qingran Yan
- Department of Rheumatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China.
| |
Collapse
|
45
|
Cantu A, Gutierrez MC, Dong X, Leek C, Anguera M, Lingappan K. Modulation of Recovery from Neonatal Hyperoxic Lung Injury by Sex as a Biological Variable. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552532. [PMID: 37609288 PMCID: PMC10441379 DOI: 10.1101/2023.08.09.552532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Recovery from lung injury during the neonatal period requires the orchestration of many biological pathways. The modulation of such pathways can drive the developing lung towards proper repair or persistent maldevelopment that can lead to a disease phenotype. Sex as a biological variable can regulate these pathways differently in the male and female lung exposed to neonatal hyperoxia. In this study, we assessed the contribution of cellular diversity in the male and female neonatal lung following injury. Our objective was to investigate sex and cell-type specific transcriptional changes that drive repair or persistent injury in the neonatal lung and delineate the alterations in the immune-endothelial cell communication networks using single cell RNA sequencing (sc-RNAseq) in a murine model of hyperoxic injury. We generated transcriptional profiles of >55,000 cells isolated from the lungs of postnatal day 1 (PND 1) and postnatal day 21 (PND 21) neonatal male and female C57BL/6 mice exposed to 95% FiO 2 between PND 1-5 (saccular stage of lung development). We show the presence of sex-based differences in the transcriptional states of lung endothelial and immune cells at PND 1 and PND 21. Furthermore, we demonstrate that biological sex significantly influences the response to injury, with a greater number of differentially expressed genes showing sex-specific patterns than those shared between male and female lungs. Pseudotime trajectory analysis highlighted genes needed for lung development that were altered by hyperoxia. Finally, we show intercellular communication between endothelial and immune cells at saccular and alveolar stages of lung development with sex-based biases in the crosstalk and identify novel ligand-receptor pairs. Our findings provide valuable insights into the cell diversity, transcriptional state, developmental trajectory, and cell-cell communication underlying neonatal lung injury, with implications for understanding lung development and possible therapeutic interventions while highlighting the crucial role of sex as a biological variable.
Collapse
|
46
|
Zou K, Zeng Z. Role of early growth response 1 in inflammation-associated lung diseases. Am J Physiol Lung Cell Mol Physiol 2023; 325:L143-L154. [PMID: 37401387 PMCID: PMC10511164 DOI: 10.1152/ajplung.00413.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/03/2023] [Accepted: 06/27/2023] [Indexed: 07/05/2023] Open
Abstract
Early growth response 1 (EGR1), which is involved in cell proliferation, differentiation, apoptosis, adhesion, migration, and immune and inflammatory responses, is a zinc finger transcription factor. EGR1 is a member of the EGR family of early response genes and can be activated by external stimuli such as neurotransmitters, cytokines, hormones, endotoxins, hypoxia, and oxidative stress. EGR1 expression is upregulated during several common respiratory diseases, such as acute lung injury/acute respiratory distress syndrome, chronic obstructive pulmonary disease, asthma, pneumonia, and novel coronavirus disease 2019. Inflammatory response is the common pathophysiological basis of these common respiratory diseases. EGR1 is highly expressed early in the disease, amplifying pathological signals from the extracellular environment and driving disease progression. Thus, EGR1 may be a target for early and effective intervention in these inflammation-associated lung diseases.
Collapse
Affiliation(s)
- Kang Zou
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical College, Ganzhou, People's Republic of China
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Zhenguo Zeng
- Department of Critical Care Medicine, Medical Center of Anesthesiology and Pain, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
47
|
Chowdary AR, Maerz T, Henn D, Hankenson KD, Pagani CA, Marini S, Gallagher K, Aguilar CA, Tower RJ, Levi B. Macrophage-mediated PDGF Activation Correlates With Regenerative Outcomes Following Musculoskeletal Trauma. Ann Surg 2023; 278:e349-e359. [PMID: 36111847 PMCID: PMC10014496 DOI: 10.1097/sla.0000000000005704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Our objective was to identify macrophage subpopulations and gene signatures associated with regenerative or fibrotic healing across different musculoskeletal injury types. BACKGROUND Subpopulations of macrophages are hypothesized to fine tune the immune response after damage, promoting either normal regenerative, or aberrant fibrotic healing. METHODS Mouse single-cell RNA sequencing data before and after injury were assembled from models of musculoskeletal injury, including regenerative and fibrotic mouse volumetric muscle loss (VML), regenerative digit tip amputation, and fibrotic heterotopic ossification. R packages Harmony , MacSpectrum , and Seurat were used for data integration, analysis, and visualizations. RESULTS There was a substantial overlap between macrophages from the regenerative VML (2 mm injury) and regenerative bone models, as well as a separate overlap between the fibrotic VML (3 mm injury) and fibrotic bone (heterotopic ossification) models. We identified 2 fibrotic-like (FL 1 and FL 2) along with 3 regenerative-like (RL 1, RL 2, and RL 3) subpopulations of macrophages, each of which was transcriptionally distinct. We found that regenerative and fibrotic conditions had similar compositions of proinflammatory and anti-inflammatory macrophages, suggesting that macrophage polarization state did not correlate with healing outcomes. Receptor/ligand analysis of macrophage-to-mesenchymal progenitor cell crosstalk showed enhanced transforming growth factor β in fibrotic conditions and enhanced platelet-derived growth factor signaling in regenerative conditions. CONCLUSION Characterization of macrophage subtypes could be used to predict fibrotic responses following injury and provide a therapeutic target to tune the healing microenvironment towards more regenerative conditions.
Collapse
Affiliation(s)
- Ashish R. Chowdary
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75235
| | - Tristan Maerz
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dominic Henn
- Department of Plastic Surgery, University of Texas Southwestern, Dallas, TX, 75235
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chase A. Pagani
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75235
| | - Simone Marini
- Department of Epidemiology, University of Florida, Gainesville, FL 32611, USA
| | - Katherine Gallagher
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carlos A. Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Robert J. Tower
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75235
| | - Benjamin Levi
- Center for Organogenesis and Trauma, Department of Surgery, University of Texas Southwestern, Dallas, TX, 75235
| |
Collapse
|
48
|
Lu H, Ma J, Li Y, Zhang J, An Y, Du W, Cai X. Bioinformatic and systems biology approach revealing the shared genes and molecular mechanisms between COVID-19 and non-alcoholic hepatitis. Front Mol Biosci 2023; 10:1164220. [PMID: 37405258 PMCID: PMC10315682 DOI: 10.3389/fmolb.2023.1164220] [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: 02/12/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023] Open
Abstract
Introduction: Coronavirus disease 2019 (COVID-19) has become a global pandemic and poses a serious threat to human health. Many studies have shown that pre-existing nonalcoholic steatohepatitis (NASH) can worsen the clinical symptoms in patients suffering from COVID-19. However, the potential molecular mechanisms between NASH and COVID-19 remain unclear. To this end, key molecules and pathways between COVID-19 and NASH were herein explored by bioinformatic analysis. Methods: The common differentially expressed genes (DEGs) between NASH and COVID-19 were obtained by differential gene analysis. Enrichment analysis and protein-protein interaction (PPI) network analysis were carried out using the obtained common DEGs. The key modules and hub genes in PPI network were obtained by using the plug-in of Cytoscape software. Subsequently, the hub genes were verified using datasets of NASH (GSE180882) and COVID-19 (GSE150316), and further evaluated by principal component analysis (PCA) and receiver operating characteristic (ROC). Finally, the verified hub genes were analyzed by single-sample gene set enrichment analysis (ssGSEA) and NetworkAnalyst was used for the analysis of transcription factor (TF)-gene interactions, TF-microRNAs (miRNA) coregulatory network, and Protein-chemical Interactions. Results: A total of 120 DEGs between NASH and COVID-19 datasets were obtained, and the PPI network was constructed. Two key modules were obtained via the PPI network, and enrichment analysis of the key modules revealed the common association between NASH and COVID-19. In total, 16 hub genes were obtained by five algorithms, and six of them, namely, Kruppel-like factor 6 (KLF6), early growth response 1 (EGR1), growth arrest and DNA-damage-inducible 45 beta (GADD45B), JUNB, FOS, and FOS-like antigen 1 (FOSL1) were confirmed to be closely related to NASH and COVID-19. Finally, the relationship between hub genes and related pathways was analyzed, and the interaction network of six hub genes was constructed with TFs, miRNAs, and compounds. Conclusion: This study identified six hub genes related to COVID-19 and NASH, providing a new perspective for disease diagnosis and drug development.
Collapse
|
49
|
Guo Y, Miao X, Sun X, Li L, Zhou A, Zhu X, Xu Y, Wang Q, Li Z, Fan Z. Zinc finger transcription factor Egf1 promotes non-alcoholic fatty liver disease. JHEP Rep 2023; 5:100724. [PMID: 37234276 PMCID: PMC10206499 DOI: 10.1016/j.jhepr.2023.100724] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/19/2023] [Accepted: 02/22/2023] [Indexed: 05/27/2023] Open
Abstract
Background & Aims Non-alcoholic fatty liver disease (NAFLD) contributes to the global epidemic of metabolic syndrome and is considered a prelude to end-stage liver diseases such as cirrhosis and hepatocellular carcinoma. During NAFLD pathogenesis, hepatic parenchymal cells (hepatocytes) undergo both morphological and functional changes owing to a rewired transcriptome. The underlying mechanism is not entirely clear. In the present study, we investigated the involvement of early growth response 1 (Egr1) in NAFLD. Methods Quantitative PCR, Western blotting, and histochemical staining were used to assess gene expression levels. Chromatin immunoprecipitation was used to evaluate protein binding to DNA. NAFLD was evaluated in leptin receptor-deficient (db/db) mice. Results We report here that Egr1 was upregulated by pro-NAFLD stimuli in vitro and in vivo. Further analysis revealed that serum response factor (SRF) was recruited to the Egr1 promoter and mediated Egr1 transactivation. Importantly, Egr1 depletion markedly mitigated NAFLD in db/db mice. RNA sequencing revealed that Egr1 knockdown in hepatocytes, on the one hand, boosted fatty acid oxidation (FAO) and, on the other hand, suppressed the synthesis of chemoattractants. Mechanistically, Egr1 interacted with peroxisome proliferator-activated receptor α (PPARα) to repress PPARα-dependent transcription of FAO genes by recruiting its co-repressor NGFI-A binding protein 1 (Nab1), which potentially led to promoter deacetylation of FAO genes. Conclusions Our data identify Egr1 as a novel modulator of NAFLD and a potential target for NAFLD intervention. Impact and Implications Non-alcoholic fatty liver disease (NAFLD) precedes cirrhosis and hepatocellular carcinoma. In this paper, we describe a novel mechanism whereby early growth response 1 (Egr1), a transcription factor, contributes to NAFLD pathogenesis by regulating fatty acid oxidation. Our data provide novel insights and translational potential for NAFLD intervention.
Collapse
Affiliation(s)
- Yan Guo
- Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Xiulian Miao
- Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Xinyue Sun
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Luyang Li
- Department of Oral Medicine, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Anqi Zhou
- Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Xi Zhu
- Department of Infectious Diseases, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Yong Xu
- Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Qinghua Wang
- Department of Gastroenterology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Zilong Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Zhiwen Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
50
|
Wang Y, He Y, Dong W, Jia M, Yang C, Wang J. DDIT3 aggravates pulpitis by modulating M1 polarization through EGR1 in macrophages. Int Immunopharmacol 2023; 120:110328. [PMID: 37235961 DOI: 10.1016/j.intimp.2023.110328] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023]
Abstract
DNA damage-inducible transcript 3 (DDIT3), a stress response gene, engages in the physiological and pathological processes of organisms, whereas its impact on pulpitis has not been defined yet. It has been demonstrated that macrophage polarization has a significant impact on inflammation. This research intends to investigate the effect of DDIT3 on the inflammation of pulpitis and macrophage polarization. C57BL/6J mice were used to model experimental pulpitis at 6, 12, 24, and 72 h after pulp exposure, with untreated mice as the control. The progression of pulpitis was visible histologically, and DDIT3 showed a trend of initially upward and downward later. Compared with wild-type mice, inflammatory cytokines and M1 macrophages were reduced, while M2 macrophages were increased in DDIT3 knockout mice. In RAW264.7 cells and bone borrow-derived macrophages, DDIT3 was found to enhance M1 polarization while impair M2 polarization. Targeted knockdown of early growth response 1 (EGR1) could rescue the blocking effect of DDIT3 deletion on M1 polarization. In conclusion, our results indicated that DDIT3 could exacerbate inflammation of pulpitis through the regulation of macrophage polarization, and DDIT3 could promote M1 polarization by inhibiting EGR1. It provides a new target for pulpitis treatment and tissue regeneration in the future.
Collapse
Affiliation(s)
- Yan Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Ying He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Wei Dong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Meie Jia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Chang Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Jiawei Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China.
| |
Collapse
|