1
|
Hoeh AE, Chang JH, Mueller RS, Basche M, Fantin A, Sepetis A, De Rossi G, Dritsoula A, Ali RR, Turowski P, Moss SE, Greenwood J. LRG1 Alters Pericyte Phenotype and Compromises Vascular Maturation. Cells 2025; 14:593. [PMID: 40277918 DOI: 10.3390/cells14080593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 04/02/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Upregulation of leucine-rich alpha-2-glycoprotein-1 (LRG1) contributes to aberrant neovascularization in many different diseases. In contrast, LRG1 is not involved in developmental angiogenesis. Here, we investigated the vasculopathic properties of LRG1 by examining its effect on developing retinal blood vessels. By injecting recombinant protein or an expression vector into the mouse retina during vascular development, we showed that exogenous LRG1 reduces pericyte coverage and NG2 expression. It leads to diminished collagen IV sheathing, fewer adhesion and gap junctions, and reduced vessel calibre and vascular density. Moreover, in mouse retinae containing exogenous LRG1, the developing blood-retinal barrier remains more permeable with significantly higher numbers of transcytotic vesicles present in microvascular endothelial cells. These results reveal that exogeneous LRG1 is sufficient to interfere with the maturation of developing retinal vessels and drive vessel development towards a dysfunctional phenotype. These observations deliver further evidence that LRG1 is an angiopathic factor and highlight the therapeutic potential of blocking LRG1 in diseases characterized by pathogenic angiogenesis or vascular remodelling.
Collapse
Affiliation(s)
- Alexandra E Hoeh
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Jui-Hsien Chang
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Ronja S Mueller
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Mark Basche
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | | | - Anastasios Sepetis
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Giulia De Rossi
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Athina Dritsoula
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Robin R Ali
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
- Ocular Cell and Gene Therapy Group, Centre for Gene Therapy and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Patric Turowski
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Stephen E Moss
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - John Greenwood
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| |
Collapse
|
2
|
Dritsoula A, Camilli C, Moss SE, Greenwood J. The disruptive role of LRG1 on the vasculature and perivascular microenvironment. Front Cardiovasc Med 2024; 11:1386177. [PMID: 38745756 PMCID: PMC11091338 DOI: 10.3389/fcvm.2024.1386177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024] Open
Abstract
The establishment of new blood vessels, and their subsequent stabilization, is a critical process that facilitates tissue growth and organ development. Once established, vessels need to diversify to meet the specific needs of the local tissue and to maintain homeostasis. These processes are tightly regulated and fundamental to normal vessel and tissue function. The mechanisms that orchestrate angiogenesis and vessel maturation have been widely studied, with signaling crosstalk between endothelium and perivascular cells being identified as an essential component. In disease, however, new vessels develop abnormally, and existing vessels lose their specialization and function, which invariably contributes to disease progression. Despite considerable research into the vasculopathic mechanisms in disease, our knowledge remains incomplete. Accordingly, the identification of angiocrine and angiopathic molecules secreted by cells within the vascular microenvironment, and their effect on vessel behaviour, remains a major research objective. Over the last decade the secreted glycoprotein leucine-rich α-2 glycoprotein 1 (LRG1), has emerged as a significant vasculopathic molecule, stimulating defective angiogenesis, and destabilizing the existing vasculature mainly, but not uniquely, by altering both canonical and non-canonical TGF-β signaling in a highly cell and context dependent manner. Whilst LRG1 does not possess any overt homeostatic role in vessel development and maintenance, growing evidence provides a compelling case for LRG1 playing a pleiotropic role in disrupting the vasculature in many disease settings. Thus, LRG1 has now been reported to damage vessels in various disorders including cancer, diabetes, chronic kidney disease, ocular disease, and lung disease and the signaling processes that drive this dysfunction are being defined. Moreover, therapeutic targeting of LRG1 has been widely proposed to re-establish a quiescent endothelium and normalized vasculature. In this review, we consider the current status of our understanding of the role of LRG1 in vascular pathology, and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Athina Dritsoula
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | | | | | | |
Collapse
|
3
|
Ma L, Wang W, Zhao Y, Liu M, Ye W, Li X. Application of LRG mechanism in normal pressure hydrocephalus. Heliyon 2024; 10:e23940. [PMID: 38223707 PMCID: PMC10784321 DOI: 10.1016/j.heliyon.2023.e23940] [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: 03/28/2023] [Revised: 11/02/2023] [Accepted: 12/15/2023] [Indexed: 01/16/2024] Open
Abstract
Normal pressure hydrocephalus (NPH) is a prevalent type of hydrocephalus, including secondary normal pressure hydrocephalus (SNPH) and idiopathic normal pressure hydrocephalus (INPH). However, its clinical diagnosis and pathological mechanism are still unclear. Leucine-rich α-2 glycoprotein (LRG) is involved in various human diseases, including cancer, diabetes, cardiovascular disease, and nervous system diseases. Now the physiological mechanism of LRG is still being explored. According to the current research results on LRG, we found that the agency of LRG has much to do with the known pathological process of NPH. This review focuses on analyzing the LRG signaling pathways and the pathological mechanism of NPH. According to the collected literature evidence, we speculated that LRG probably be involved in the pathological process of NPH. Finally, based on the mechanism of LRG and NPH, we also summarized the evidence of molecular targeted therapies for future research and clinical application.
Collapse
Affiliation(s)
| | | | - Yongqiang Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Menghao Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Wei Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xianfeng Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
4
|
Cuarental L, Ribagorda M, Ceballos MI, Pintor-Chocano A, Carriazo SM, Dopazo A, Vazquez E, Suarez-Alvarez B, Cannata-Ortiz P, Sanz AB, Ortiz A, Sanchez-Niño MD. The transcription factor Fosl1 preserves Klotho expression and protects from acute kidney injury. Kidney Int 2023; 103:686-701. [PMID: 36565807 DOI: 10.1016/j.kint.2022.11.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 11/02/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022]
Abstract
Increased expression of AP-1 transcription factor components has been reported in acute kidney injury (AKI). However, the role of specific components, such as Fosl1, in tubular cells or AKI is unknown. Upstream regulator analysis of murine nephrotoxic AKI transcriptomics identified AP-1 as highly upregulated. Among AP-1 canonical components, Fosl1 was found to be upregulated in two transcriptomics datasets from nephrotoxic murine AKI induced by folic acid or cisplatin and from proximal tubular cells exposed to TWEAK, a cytokine mediator of AKI. Fosl1 was minimally expressed in the kidneys of control uninjured mice. Increased Fosl1 protein was localized to proximal tubular cell nuclei in AKI. In human AKI, FOSL1 was found present in proximal tubular cells in kidney sections and in urine along with increased urinary FOSL1 mRNA. Selective Fosl1 deficiency in proximal tubular cells (Fosl1Δtub) increased the severity of murine cisplatin- or folate-induced AKI as characterized by lower kidney function, more severe kidney inflammation and Klotho downregulation. Indeed, elevated AP-1 activity was observed after cisplatin-induced AKI in Fosl1Δtub mice compared to wild-type mice. More severe Klotho downregulation preceded more severe kidney dysfunction. The Klotho promoter was enriched in Fosl1 binding sites and Fosl1 bound to the Klotho promoter in cisplatin-AKI. In cultured proximal tubular cells, Fosl1 targeting increased the proinflammatory response and downregulated Klotho. In vivo, recombinant Klotho administration protected Fosl1Δtub mice from cisplatin-AKI. Thus, increased proximal tubular Fosl1 expression during AKI is an adaptive response, preserves Klotho, and limits the severity of tubular cell injury and AKI.
Collapse
Affiliation(s)
- Leticia Cuarental
- Department of Nephrology and Hypertension, Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain; RICORS2040 (Redes de Investigación Cooperativa Orientadas a Resultados en Salud), Madrid, Spain
| | - Marta Ribagorda
- Department of Nephrology and Hypertension, Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain; RICORS2040 (Redes de Investigación Cooperativa Orientadas a Resultados en Salud), Madrid, Spain
| | - Maria I Ceballos
- Department of Nephrology and Hypertension, Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain; RICORS2040 (Redes de Investigación Cooperativa Orientadas a Resultados en Salud), Madrid, Spain
| | - Aranzazu Pintor-Chocano
- Department of Nephrology and Hypertension, Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain; RICORS2040 (Redes de Investigación Cooperativa Orientadas a Resultados en Salud), Madrid, Spain
| | - Sol M Carriazo
- Department of Nephrology and Hypertension, Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain; RICORS2040 (Redes de Investigación Cooperativa Orientadas a Resultados en Salud), Madrid, Spain
| | - Ana Dopazo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Enrique Vazquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz Suarez-Alvarez
- Translational Immunology, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Pablo Cannata-Ortiz
- Department of Pathology, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
| | - Ana B Sanz
- Department of Nephrology and Hypertension, Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain; RICORS2040 (Redes de Investigación Cooperativa Orientadas a Resultados en Salud), Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain; RICORS2040 (Redes de Investigación Cooperativa Orientadas a Resultados en Salud), Madrid, Spain; Departamento de Medicina, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Maria D Sanchez-Niño
- Department of Nephrology and Hypertension, Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain; RICORS2040 (Redes de Investigación Cooperativa Orientadas a Resultados en Salud), Madrid, Spain; Departamento de Farmacología, Universidad Autonoma de Madrid (UAM), Madrid, Spain.
| |
Collapse
|
5
|
Influence of FOSL1 Inhibition on Vascular Calcification and ROS Generation through Ferroptosis via P53-SLC7A11 Axis. Biomedicines 2023; 11:biomedicines11020635. [PMID: 36831172 PMCID: PMC9953509 DOI: 10.3390/biomedicines11020635] [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: 01/14/2023] [Revised: 02/04/2023] [Accepted: 02/13/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Vascular calcification during aging is highly prevalent in patients with cardiovascular disease; however, there is still no improvement in clarifying the development of vascular calcification. FOSL1 is a transcription regulator belonging to the AP-1 family, which has a unique function in vascular senescence, but its role in vascular calcification needs to be further explored. METHODS Primary mouse vascular smooth muscle cells were isolated and used to construct a calcification model in vitro. Seven-week-old male C57BL/6 mice were used to build the vitD3-induced calcification model in vivo. qRT-PCR and western blot were used to verify the expression of FOSL1 and other genes expressed in vascular smooth muscle cells and aortas. The level of calcification was determined by Alizarin Red S (ARS) staining and the calcium content assay. The level of cellular GSH was detected by the GSH assay kit. RESULTS Here, we report that FOSL1 was up-regulated after high-calcium/phosphate treatment in both the in vivo and in vitro vascular calcification models. Functional studies have shown that the reduction of FOSL1 attenuates ferroptosis and calcification in vascular smooth muscle cells, as indicated by ARS staining, calcium content assay, and western blot. The inhibition of FOSL1 downregulated the expression of bone-related molecules including Msh Homeobox 2 (MSX2) and tumor necrosis factor receptor superfamily, member 11b/osteoprotegerin (OPG), suggesting that FOSL1 promoted osteogenic differentiation of vascular smooth muscle cells. Furthermore, we found that the ferroptosis-inducing drug erastin can significantly accelerate calcification in the aortic ring while Ferrostatin-1 (fer-1), a drug to protect cells from ferroptosis, can alleviate calcification. Further experiments have shown that inhibiting FOSL1 can promote the expression of ferroptosis-related genes and attenuate calcification. Functionally, cellular GSH levels were increased after the reduction of FOSL1. CONCLUSIONS In this study, we observed a significant protective effect when we reduced the expression of FOSL1 during vascular calcification, and this effect might regulate ferroptosis to a great extent.
Collapse
|
6
|
Xia S, Vila Ellis L, Winkley K, Menden H, Mabry SM, Venkatraman A, Louiselle D, Gibson M, Grundberg E, Chen J, Sampath V. Neonatal hyperoxia induces activated pulmonary cellular states and sex-dependent transcriptomic changes in a model of experimental bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2023; 324:L123-L140. [PMID: 36537711 PMCID: PMC9902224 DOI: 10.1152/ajplung.00252.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
Hyperoxia disrupts lung development in mice and causes bronchopulmonary dysplasia (BPD) in neonates. To investigate sex-dependent molecular and cellular programming involved in hyperoxia, we surveyed the mouse lung using single cell RNA sequencing (scRNA-seq), and validated our findings in human neonatal lung cells in vitro. Hyperoxia-induced inflammation in alveolar type (AT) 2 cells gave rise to damage-associated transient progenitors (DATPs). It also induced a new subpopulation of AT1 cells with reduced expression of growth factors normally secreted by AT1 cells, but increased mitochondrial gene expression. Female alveolar epithelial cells had less EMT and pulmonary fibrosis signaling in hyperoxia. In the endothelium, expansion of Car4+ EC (Cap2) was seen in hyperoxia along with an emergent subpopulation of Cap2 with repressed VEGF signaling. This regenerative response was increased in females exposed to hyperoxia. Mesenchymal cells had inflammatory signatures in hyperoxia, with a new distal interstitial fibroblast subcluster characterized by repressed lipid biosynthesis and a transcriptomic signature resembling myofibroblasts. Hyperoxia-induced gene expression signatures in human neonatal fibroblasts and alveolar epithelial cells in vitro resembled mouse scRNA-seq data. These findings suggest that neonatal exposure to hyperoxia programs distinct sex-specific stem cell progenitor and cellular reparative responses that underpin lung remodeling in BPD.
Collapse
Affiliation(s)
- Sheng Xia
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Lisandra Vila Ellis
- Department of Pulmonary Medicine, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Konner Winkley
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Heather Menden
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Sherry M Mabry
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Aparna Venkatraman
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Daniel Louiselle
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Margaret Gibson
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Elin Grundberg
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
- Children's Mercy Research Institute, Kansas City, Missouri
| | - Jichao Chen
- Department of Pulmonary Medicine, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Venkatesh Sampath
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
- Children's Mercy Research Institute, Kansas City, Missouri
| |
Collapse
|
7
|
Otálora-Otálora BA, López-Kleine L, Rojas A. Lung Cancer Gene Regulatory Network of Transcription Factors Related to the Hallmarks of Cancer. Curr Issues Mol Biol 2023; 45:434-464. [PMID: 36661515 PMCID: PMC9857713 DOI: 10.3390/cimb45010029] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 01/06/2023] Open
Abstract
The transcriptomic analysis of microarray and RNA-Seq datasets followed our own bioinformatic pipeline to identify a transcriptional regulatory network of lung cancer. Twenty-six transcription factors are dysregulated and co-expressed in most of the lung cancer and pulmonary arterial hypertension datasets, which makes them the most frequently dysregulated transcription factors. Co-expression, gene regulatory, coregulatory, and transcriptional regulatory networks, along with fibration symmetries, were constructed to identify common connection patterns, alignments, main regulators, and target genes in order to analyze transcription factor complex formation, as well as its synchronized co-expression patterns in every type of lung cancer. The regulatory function of the most frequently dysregulated transcription factors over lung cancer deregulated genes was validated with ChEA3 enrichment analysis. A Kaplan-Meier plotter analysis linked the dysregulation of the top transcription factors with lung cancer patients' survival. Our results indicate that lung cancer has unique and common deregulated genes and transcription factors with pulmonary arterial hypertension, co-expressed and regulated in a coordinated and cooperative manner by the transcriptional regulatory network that might be associated with critical biological processes and signaling pathways related to the acquisition of the hallmarks of cancer, making them potentially relevant tumor biomarkers for lung cancer early diagnosis and targets for the development of personalized therapies against lung cancer.
Collapse
Affiliation(s)
- Beatriz Andrea Otálora-Otálora
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá 110131, Colombia
- Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 11001, Colombia
| | - Liliana López-Kleine
- Departamento de Estadística, Universidad Nacional de Colombia, Bogotá 11001, Colombia
- Correspondence: (L.L.-K.); (A.R.)
| | - Adriana Rojas
- Facultad de Medicina, Instituto de Genética Humana, Pontificia Universidad Javeriana, Bogotá 110211, Colombia
- Correspondence: (L.L.-K.); (A.R.)
| |
Collapse
|
8
|
Lipopolysaccharide-Induced Immunological Tolerance in Monocyte-Derived Dendritic Cells. IMMUNO 2022. [DOI: 10.3390/immuno2030030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Bacterial lipopolysaccharides (LPS), also referred to as endotoxins, are major outer surface membrane components present on almost all Gram-negative bacteria and are major determinants of sepsis-related clinical complications including septic shock. LPS acts as a strong stimulator of innate or natural immunity in a wide variety of eukaryotic species ranging from insects to humans including specific effects on the adaptive immune system. However, following immune stimulation, lipopolysaccharide can induce tolerance which is an essential immune-homeostatic response that prevents overactivation of the inflammatory response. The tolerance induced by LPS is a state of reduced immune responsiveness due to persistent and repeated challenges, resulting in decreased expression of pro-inflammatory modulators and up-regulation of antimicrobials and other mediators that promote a reduction of inflammation. The presence of environmental-derived LPS may play a key role in decreasing autoimmune diseases and gut tolerance to the plethora of ingested antigens. The use of LPS may be an important immune adjuvant as demonstrated by the promotion of IDO1 increase when present in the fusion protein complex of CTB-INS (a chimera of the cholera toxin B subunit linked to proinsulin) that inhibits human monocyte-derived DC (moDC) activation, which may act through an IDO1-dependent pathway. The resultant state of DC tolerance can be further enhanced by the presence of residual E. coli lipopolysaccharide (LPS) which is almost always present in partially purified CTB-INS preparations. The approach to using an adjuvant with an autoantigen in immunotherapy promises effective treatment for devastating tissue-specific autoimmune diseases like multiple sclerosis (MS) and type 1 diabetes (T1D).
Collapse
|
9
|
He YY, Zhou HF, Chen L, Wang YT, Xie WL, Xu ZZ, Xiong Y, Feng YQ, Liu GY, Li X, Liu J, Wu QP. The Fra-1: Novel role in regulating extensive immune cell states and affecting inflammatory diseases. Front Immunol 2022; 13:954744. [PMID: 36032067 PMCID: PMC9404335 DOI: 10.3389/fimmu.2022.954744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Fra-1(Fos-related antigen1), a member of transcription factor activator protein (AP-1), plays an important role in cell proliferation, apoptosis, differentiation, inflammation, oncogenesis and tumor metastasis. Accumulating evidence suggest that the malignancy and invasive ability of tumors can be significantly changed by directly targeting Fra-1. Besides, the effects of Fra-1 are gradually revealed in immune and inflammatory settings, such as arthritis, pneumonia, psoriasis and cardiovascular disease. These regulatory mechanisms that orchestrate immune and non-immune cells underlie Fra-1 as a potential therapeutic target for a variety of human diseases. In this review, we focus on the current knowledge of Fra-1 in immune system, highlighting its unique importance in regulating tissue homeostasis. In addition, we also discuss the possible critical intervention strategy in diseases, which also outline future research and development avenues.
Collapse
|
10
|
Regulation of Inflammation-Related Genes through Fosl1 Suppression in a Levetiracetam-Treated Pilocarpine-Induced Status Epilepticus Mouse Model. Int J Mol Sci 2022; 23:ijms23147608. [PMID: 35886955 PMCID: PMC9317701 DOI: 10.3390/ijms23147608] [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: 06/06/2022] [Revised: 06/28/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Levetiracetam (LEV) suppresses the upregulation of proinflammatory molecules that occurs during epileptogenesis after status epilepticus (SE). Based on previous studies, LEV likely helps prevent the onset of epilepsy after insults to the brain, unlike other conventional anti-epileptic drugs. Recently, we discovered that the increase in Fosl1 expression that occurs after lipopolysaccharide (LPS) stimulation is suppressed by LEV and that Fosl1 inhibition suppresses inflammation in BV-2 microglial cells. These data indicate that Fosl1 is an important target of LEV and a key factor in preventing epilepsy onset. In this study, we examined the effects of LEV on Fosl1 expression and neuroinflammation in vivo. During epileptogenesis, the post-SE upregulation of hippocampal levels of Fosl1 and many inflammatory factors were suppressed by LEV. Fosl1 expression showed a characteristic pattern different from that of the expression of Fos, an immediate-early gene belonging to the same Fos family. At 2 days after SE, Fosl1 was predominantly expressed in astrocytes but was rarely detected in microglia, whereas Fos expression was distributed in various brain cell types. The expression of A2 astrocyte markers was similar to that of Fosl1 and was significantly suppressed by LEV. These results suggest that LEV may regulate astrocyte reactivity through regulation of Fosl1.
Collapse
|
11
|
He W, Zhou X, Mao Y, Wu Y, Tang X, Yan S, Tang S. CircCRIM1 promotes nasopharyngeal carcinoma progression via the miR-34c-5p/FOSL1 axis. Eur J Med Res 2022; 27:59. [PMID: 35484574 PMCID: PMC9052594 DOI: 10.1186/s40001-022-00667-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a rare malignancy with multiple risk factors (Epstein-Barr virus, etc.) that seriously threatens the health of people. CircRNAs are known to regulate the tumorigenesis of malignant tumours, including NPC. Moreover, circCRIM1 expression is reported to be upregulated in NPC. Nevertheless, the impact of circCRIM1 on NPC progression is not clear. METHODS An MTT assay was performed to assess cell viability. In addition, cell invasion and migration were assessed by the transwell assay. Dual luciferase assays were performed to assess the association among circCRIM1, miR-34c-5p and FOSL1. Moreover, RT-qPCR was applied to assess mRNA levels, and protein levels were determined by Western blot. RESULTS CircCRIM1 and FOSL1 were upregulated in NPC cells, while miR-34c-5p was downregulated. Knockdown of circCRIM1 significantly decreased the invasion, viability and migration of NPC cells. The miR-34c-5p inhibitor notably promoted the malignant behaviour of NPC cells, while miR-34c-5p mimics exerted the opposite effect. Moreover, circCRIM1 could bind with miR-34c-5p, and FOSL1 was identified to be downstream of miR-34c-5p. Furthermore, circCRIM1 downregulation notably inhibited the proliferation and invasion of NPC cells, while this phenomenon was significantly reversed by FOSL1 overexpression. CONCLUSION Silencing circCRIM1 inhibited the tumorigenesis of NPC. Thus, circCRIM1 might be a novel target for NPC.
Collapse
Affiliation(s)
- Weifeng He
- Oncology Department, The Second People's Hospital of Hunan Province, Changsha, 410007, Hunan, People's Republic of China
| | - Xiangqi Zhou
- Oncology Department, Affiliated Nanhua Hospital of University of South China, No. 336 Dong Feng South Road, Hengyang, 421002, Hunan, People's Republic of China
| | - Yini Mao
- Oncology Department, The Second People's Hospital of Hunan Province, Changsha, 410007, Hunan, People's Republic of China
| | - YangJie Wu
- Oncology Department, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Xiyang Tang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Sijia Yan
- Oncology Department, Affiliated Nanhua Hospital of University of South China, No. 336 Dong Feng South Road, Hengyang, 421002, Hunan, People's Republic of China.
| | - Sanyuan Tang
- Oncology Department, The Second People's Hospital of Hunan Province, Changsha, 410007, Hunan, People's Republic of China. .,Oncology Department, Affiliated Nanhua Hospital of University of South China, No. 336 Dong Feng South Road, Hengyang, 421002, Hunan, People's Republic of China.
| |
Collapse
|
12
|
Akangire G, Menden H, Xia S, Thiffault I, Ahmed A, Sampath V. EPHB4 Mutation Suppresses PROX1 Expression and Disrupts Lymphatic Development in Neonatal Hydrops. Pediatrics 2022; 149:184852. [PMID: 35178555 DOI: 10.1542/peds.2021-053294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/29/2021] [Indexed: 11/24/2022] Open
Abstract
This case report highlights the importance of screening for mutations in EPHB4 and other genes that regulate lymphatic development in infants with the nonimmune hydrops fetalis.
Collapse
Affiliation(s)
- Gangaram Akangire
- Division of Neonatology.,University of Missouri Kansas City-Kansas City, Kansas City, Missouri
| | | | | | - Isabelle Thiffault
- Center for Genomic Medicine.,Department of Pathology and Laboratory Medicine.,University of Missouri Kansas City-Kansas City, Kansas City, Missouri
| | - Atif Ahmed
- Department of Pathology, Children's Mercy Hospital
| | - Venkatesh Sampath
- Division of Neonatology.,University of Missouri Kansas City-Kansas City, Kansas City, Missouri
| |
Collapse
|
13
|
Zou Y, Xu Y, Chen X, Wu Y, Fu L, Lv Y. Research Progress on Leucine-Rich Alpha-2 Glycoprotein 1: A Review. Front Pharmacol 2022; 12:809225. [PMID: 35095520 PMCID: PMC8797156 DOI: 10.3389/fphar.2021.809225] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/13/2021] [Indexed: 12/18/2022] Open
Abstract
Leucine-rich alpha⁃2 glycoprotein 1 (LRG1) is an important member of the leucine-rich repetitive sequence protein family. LRG1 was mainly involved in normal physiological activities of the nervous system, such as synapse formation, synapse growth, the development of nerve processes, neurotransmitter transfer and release, and cell adhesion molecules or ligand-binding proteins. Also, LRG1 affected the development of respiratory diseases, hematological diseases, endocrine diseases, tumor diseases, eye diseases, cardiovascular diseases, rheumatic immune diseases, infectious diseases, etc. LRG1 was a newly discovered important upstream signaling molecule of transforming growth factor⁃β (TGF⁃β) that affected various pathological processes through the TGF⁃β signaling pathway. However, research on LRG1 and its involvement in the occurrence and development of diseases was still in its infancy and the current studies were mainly focused on proteomic detection and basic animal experimental reports. We could reasonably predict that LRG1 might act as a new direction and strategy for the treatment of many diseases.
Collapse
Affiliation(s)
- Yonghui Zou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China.,School of Clinical Medicine, Nanchang University, Nanchang, China
| | - Yi Xu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China.,School of Clinical Medicine, Nanchang University, Nanchang, China
| | - Xiaofeng Chen
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China.,School of Clinical Medicine, Nanchang University, Nanchang, China
| | - Yaoqi Wu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China.,College of Pharmacy, Nanchang University, Nanchang, China
| | - Longsheng Fu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yanni Lv
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
14
|
Camilli C, Hoeh AE, De Rossi G, Moss SE, Greenwood J. LRG1: an emerging player in disease pathogenesis. J Biomed Sci 2022; 29:6. [PMID: 35062948 PMCID: PMC8781713 DOI: 10.1186/s12929-022-00790-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/11/2022] [Indexed: 12/15/2022] Open
Abstract
The secreted glycoprotein leucine-rich α-2 glycoprotein 1 (LRG1) was first described as a key player in pathogenic ocular neovascularization almost a decade ago. Since then, an increasing number of publications have reported the involvement of LRG1 in multiple human conditions including cancer, diabetes, cardiovascular disease, neurological disease, and inflammatory disorders. The purpose of this review is to provide, for the first time, a comprehensive overview of the LRG1 literature considering its role in health and disease. Although LRG1 is constitutively expressed by hepatocytes and neutrophils, Lrg1-/- mice show no overt phenotypic abnormality suggesting that LRG1 is essentially redundant in development and homeostasis. However, emerging data are challenging this view by suggesting a novel role for LRG1 in innate immunity and preservation of tissue integrity. While our understanding of beneficial LRG1 functions in physiology remains limited, a consistent body of evidence shows that, in response to various inflammatory stimuli, LRG1 expression is induced and directly contributes to disease pathogenesis. Its potential role as a biomarker for the diagnosis, prognosis and monitoring of multiple conditions is widely discussed while dissecting the mechanisms underlying LRG1 pathogenic functions. Emphasis is given to the role that LRG1 plays as a vasculopathic factor where it disrupts the cellular interactions normally required for the formation and maintenance of mature vessels, thereby indirectly contributing to the establishment of a highly hypoxic and immunosuppressive microenvironment. In addition, LRG1 has also been reported to affect other cell types (including epithelial, immune, mesenchymal and cancer cells) mostly by modulating the TGFβ signalling pathway in a context-dependent manner. Crucially, animal studies have shown that LRG1 inhibition, through gene deletion or a function-blocking antibody, is sufficient to attenuate disease progression. In view of this, and taking into consideration its role as an upstream modifier of TGFβ signalling, LRG1 is suggested as a potentially important therapeutic target. While further investigations are needed to fill gaps in our current understanding of LRG1 function, the studies reviewed here confirm LRG1 as a pleiotropic and pathogenic signalling molecule providing a strong rationale for its use in the clinic as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Carlotta Camilli
- Institute of Ophthalmology, University College London, London, UK.
| | - Alexandra E Hoeh
- Institute of Ophthalmology, University College London, London, UK
| | - Giulia De Rossi
- Institute of Ophthalmology, University College London, London, UK
| | - Stephen E Moss
- Institute of Ophthalmology, University College London, London, UK
| | - John Greenwood
- Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
15
|
Kawai Y, Hayakawa M, Tanaka T, Yamada Y, Nakayama A, Kato Y, Kouwaki M, Kato T, Tanaka R, Muramatsu K, Hayashi S, Yamamoto H, Takemoto K, Ieda K, Nagaya Y, Honda S, Shinohara O, Funato Y, Kokubo M, Imamine H, Miyata M. Pulmonary hypertension with bronchopulmonary dysplasia: Aichi cohort study. Pediatr Int 2022; 64:e15271. [PMID: 35972055 DOI: 10.1111/ped.15271] [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: 02/02/2022] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The incidence of pulmonary hypertension (PH) associated with bronchopulmonary dysplasia (BPD) has not been investigated in regional cohorts. The aim of this study was to clarify the incidence of PH associated with BPD in all very low birthweight infants (VLBWIs) born during the study period in Aichi Prefecture, Japan. METHODS We conducted a retrospective observational cohort study of all VLBWIs born in Aichi Prefecture. The inclusion criteria were VLB, birth between 1 January 2015 and 31 December 2015, and admission to any neonatal intensive care unit in Aichi Prefecture. BPD28d and BPD36w were defined as the need for supplemental oxygen or any respiratory support at 28 days of age or 36 weeks of postmenstrual age (PMA). The primary outcome was the incidence of PH after 36 weeks' PMA (PH36w) in VLBWIs with BPD28d and BPD36w. The secondary outcomes were the clinical factors related to PH36w in BPD36w patients. Mann-Whitney U-test and Fisher's exact test were used for univariate analysis. Differences were considered statistically significant at P < 0.05. Risk ratio (RR) and 95% confidence interval (CI) were also evaluated. RESULTS A total of 441 patients were analyzed. A total of 217 and 131 patients met the definition of BPD28d and BPD36w, respectively. Nine patients were diagnosed with PH36w (4.2% and 6.9% of the BPD28d and BPD36w patients, respectively). The presence of oligohydramnios (RR, 2.71; 95% CI: 1.55-4.73, P = 0.014) and sepsis (RR, 3.62; 95% CI: 1.51-8.63, P = 0.025) was significant in the PH36w patients. CONCLUSIONS The incidence of PH36w was 4.2% and 6.9% in the BPD28d and BPD36w patients, respectively. Oligohydramnios and sepsis were significantly associated with PH36w in VLBWIs.
Collapse
Affiliation(s)
- Yuri Kawai
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Japan
| | - Masahiro Hayakawa
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Taihei Tanaka
- Department of Pediatrics, Japanese Red Cross Nagoya Daini Hospital, Nagoya, Japan
| | - Yasumasa Yamada
- Department of Perinatal and Neonatal Medicine, Aichi Medical University, Nagakute, Japan
| | - Atsushi Nakayama
- Department of Pediatrics, Japanese Red Cross Nagoya Daiichi Hospital, Nagoya, Japan
| | - Yuichi Kato
- Department of Pediatrics, Anjo Kosei Hospital, Anjo, Japan
| | - Masanori Kouwaki
- Department of Pediatrics, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Takenori Kato
- Department of Pediatrics, Toyohashi Municipal Hospital, Toyohashi, Japan.,Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ryo Tanaka
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan.,Department of Pediatrics, Japanese Red Cross Nagoya Daiichi Hospital, Nagoya, Japan
| | - Kanji Muramatsu
- Department of Pediatrics, Toyohashi Municipal Hospital, Toyohashi, Japan.,Department of Pediatrics, Nagoya City West Medical Center, Nagoya, Japan
| | - Seiji Hayashi
- Department of Pediatrics, Okazaki City Hospital, Okazaki, Japan
| | - Hikaru Yamamoto
- Department of Pediatrics, Toyota Memorial Hospital, Toyota, Japan
| | - Koji Takemoto
- Department of Pediatrics, Konan Kosei Hospital, Konan, Japan
| | - Kuniko Ieda
- Department of Pediatrics, Tosei General Hospital, Seto, Japan
| | - Yoshiaki Nagaya
- Department of Pediatrics, Ichinomiya Municipal Hospital, Ichinomiya, Japan
| | - Shigeru Honda
- Department of Pediatrics, Komaki City Hospital, Komaki, Japan
| | | | - Yusuke Funato
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Japan.,Department of Pediatrics, Kariya Toyota General Hospital, Kariya, Japan
| | - Minoru Kokubo
- Department of Pediatrics, Kainan Hospital, Yatomi, Japan
| | - Hiroki Imamine
- Department of Pediatrics, Holy Spirit Hospital, Nagoya, Japan
| | - Masafumi Miyata
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
16
|
Salimi U, Dummula K, Tucker MH, Dela Cruz CS, Sampath V. Postnatal Sepsis and Bronchopulmonary Dysplasia in Premature Infants: Mechanistic Insights into "New BPD". Am J Respir Cell Mol Biol 2021; 66:137-145. [PMID: 34644520 DOI: 10.1165/rcmb.2021-0353ps] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a debilitating disease in premature infants resulting from lung injury that disrupts alveolar and pulmonary vascular development. Despite the use of lung-protective ventilation and targeted oxygen therapy, BPD rates have not significantly changed over the last decade. Recent evidence suggests that sepsis and conditions initiating the systemic inflammatory response syndrome in preterm infants are key risk factors for BPD. However, the mechanisms by which sepsis-associated systemic inflammation and microbial dissemination program aberrant lung development are not fully understood. Progress has been made within the last 5 years with the inception of animal models allowing mechanistic investigations into neonatal acute lung injury and alveolar remodeling due to endotoxemia and NEC. These recent studies begin to unravel the pathophysiology of early endothelial immune activation via pattern recognition receptors such as Toll Like Receptor 4 and disruption of critical lung developmental processes such as angiogenesis, extracellular matrix deposition, and ultimately alveologenesis. Here we review scientific evidence from preclinical models of neonatal sepsis-induced lung injury to new data emerging from clinical literature.
Collapse
Affiliation(s)
- Umar Salimi
- Yale University, 5755, Pediatrics, New Haven, Connecticut, United States
| | - Krishna Dummula
- Children's Mercy, 4204, Pediatrics, Kansas City, Missouri, United States
| | - Megan H Tucker
- Children's Mercy, 4204, Pediatrics, Kansas City, Missouri, United States
| | - Charles S Dela Cruz
- Yale University, Pulmonary and Critical Care Medicine, New Haven, Connecticut, United States
| | - Venkatesh Sampath
- Children\'s Mercy Hospitals and Clinics, 4204, Pediatrics, Kansas City, Missouri, United States;
| |
Collapse
|
17
|
Saikosaponin-D Alleviates Renal Inflammation and Cell Apoptosis in a Mouse Model of Sepsis via TCF7/FOSL1/MMP9 Inhibition. Mol Cell Biol 2021; 41:e0033221. [PMID: 34309413 DOI: 10.1128/mcb.00332-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Evidence exists reporting that Saikosaponin-d can prevent experimental sepsis, and this study aims to illustrate the molecular events underlying its renoprotective effects on lipopolysaccharide (LPS)-induced renal inflammation simulating sepsis. Through network pharmacology analysis and bioinformatics analysis, we identified that saikosaponin-d may influence sepsis development by mediating TCF7. Dual luciferase reporter gene and ChIP assays were used to explore the interactions between TCF7, FOSL1 and MMP9. The experimental data suggested that Saikosaponin-d attenuated LPS-induced renal injury, as evidenced by reduced the production of proinflammatory cytokines as well as cell apoptosis in the renal tissues of LPS-induced mice. Mechanically, Saikosaponin-d inhibited FOSL1 by inhibiting TCF7, which reduced the expression of inflammatory factors in renal cells. TCF7 activated the FOSL1 expression and consequently promoted the expression of MMP9. Also, Saikosaponin-d reduced cell apoptosis and the expression of inflammatory factors by inhibiting the TCF7/FOSL1/MMP9 axis in vivo. In conclusion, Saikosaponin-d suppresses FOSL1 transcription by downregulating TCF7, thereby inhibiting MMP9 expression and ultimately reducing the renal inflammation and cell apoptosis induced by sepsis.
Collapse
|
18
|
Xia S, Yu W, Menden H, Younger ST, Sampath V. FOXC2 Autoregulates Its Expression in the Pulmonary Endothelium After Endotoxin Stimulation in a Histone Acetylation-Dependent Manner. Front Cell Dev Biol 2021; 9:657662. [PMID: 34017833 PMCID: PMC8129010 DOI: 10.3389/fcell.2021.657662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 04/12/2021] [Indexed: 12/26/2022] Open
Abstract
The innate immune response of pulmonary endothelial cells (EC) to lipopolysaccharide (LPS) induces Forkhead box protein C2 (FOXC2) activation through Toll Like Receptor 4 (TLR4). The mechanisms by which FOXC2 expression is regulated in lung EC under LPS stimulation remain unclear. We postulated that FOXC2 regulates its own expression in sepsis, and its transcriptional autoregulation directs lymphatic EC cell-fate decision. Bioinformatic analysis identified potential FOXC2 binding sites in the FOXC2 promoter. In human lung EC, we verified using chromatin immunoprecipitation (ChIP) and luciferase assays that FOXC2 bound to its own promoter and stimulated its expression after LPS stimulation. Chemical inhibition of histone acetylation by garcinol repressed LPS-induced histone acetylation in the FOXC2 promoter region, and disrupted LPS-mediated FOXC2 binding and transcriptional activation. CRISPR/dCas9/gRNA directed against FOXC2-binding-element (FBE) suppressed LPS-stimulated FOXC2 binding and autoregulation by blocking FBEs in the FOXC2 promoter, and repressed expression of lymphatic EC markers. In a neonatal mouse model of sterile sepsis, LPS-induced FOXC2 binding to FBE and FOXC2 expression in lung EC was attenuated with garcinol treatment. These data reveal a new mechanism of LPS-induced histone acetylation-dependent FOXC2 autoregulation.
Collapse
Affiliation(s)
- Sheng Xia
- Department of Pediatrics, Children's Mercy Kansas City, MO, United States
| | - Wei Yu
- Department of Pediatrics, Children's Mercy Kansas City, MO, United States
| | - Heather Menden
- Department of Pediatrics, Children's Mercy Kansas City, MO, United States
| | - Scott T Younger
- Center for Pediatric Genomic Medicine, Children's Mercy Kansas City, MO, United States
| | - Venkatesh Sampath
- Department of Pediatrics, Children's Mercy Kansas City, MO, United States
| |
Collapse
|
19
|
Gao J, Wu M, Wang F, Jiang L, Tian R, Zhu X, He S. CD74, a novel predictor for bronchopulmonary dysplasia in preterm infants. Medicine (Baltimore) 2020; 99:e23477. [PMID: 33235138 PMCID: PMC7710202 DOI: 10.1097/md.0000000000023477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) remains a major complication and accounts for high morbidity and mortality of preterm infants. The present study aimed to identify the key genes in the development of BPD and to provide some new insights into the pathogenesis of BPD. The GSE108754 dataset was downloaded from Gene Expression Omnibus database containing 5 samples of BPD patients and 6 of non-BPD infants. The differentially expressed genes (DEGs) between BPD and non-BPD patients were identified by R software. The pathway and function enrichment analyses were performed through Database for Annotation Visualization and Integrated Discovery website. The protein-protein interaction network for DEGs was established by Cytoscape software and the most highly connected module was selected through MCODE plugin. Furthermore, the clinical sample verification among 25 BPD patients and 10 non-BPD infants was carried out in our center. Finally, based on the results above, the gene set enrichment analysis focusing on CD74 upregulated status was employed. Totally, 189 DEGs including 147 upregulated genes and 42 downregulated genes between BPD and non-BPD patients were screened out. The pathway and function enrichments revealed these DEGs were mainly enriched in asthma, intestinal immune network for IgA production, antigen processing and presentation and immune response. Thirteen DEGs (CD74, HLA-DMA, HLA-DRA, HLA-DMB, HLA-DOB, HLA-DQA1, HLA-DRB5, HLA-DPA1, HLA-DOA, HLA-DPB1, HLA-DQB2, HLA-DQA2, and HLA-DQB1) were determined as hub genes. The mRNA expression levels of the 13 hub genes were tested by quantitative real-time polymerase chain reaction among our clinical samples. Eventually, CD74 was confirmed to be the most significant highly expressed in BPD samples (P < .001) and its expression level was negatively correlated with gestational age (r = -0.653) and birth weight (r = -0.675). The gene set enrichment analysis results showed the gene sets associated with lupus erythematosus, viral myocarditis, immune network for IgA production, graft versus host disease, cell adhesion molecules and so no were differentially enriched with the phenotype of high-expression CD74. In conclusion, CD74 may serve to predict the BPD development and provide a new therapeutic target for BPD.
Collapse
Affiliation(s)
- Junyan Gao
- Department of Pediatrics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu
| | - Mingfu Wu
- Department of Pediatrics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu
| | - Fudong Wang
- Department of Pediatrics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu
| | - Lijun Jiang
- Department of Pediatrics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu
| | - Rui Tian
- Department of Pediatrics, The First People's Hospital of Kunming City, Kunming, Yunnan
| | - Xueping Zhu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu
| | - Shan He
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu
- Department of Pediatrics, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| |
Collapse
|