1
|
Palomino-Vizcaino G, Bañuelos-Villegas EG, Alvarez-Salas LM. The Natural History of Cervical Cancer and the Case for MicroRNAs: Is Human Papillomavirus Infection the Whole Story? Int J Mol Sci 2024; 25:12991. [PMID: 39684702 PMCID: PMC11641362 DOI: 10.3390/ijms252312991] [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: 11/18/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs (ncRNAs) that negatively regulate gene expression. MiRNAs regulate fundamental biological processes and have significant roles in several pathologies, including cancer. Cervical cancer is the best-known example of a widespread human malignancy with a demonstrated viral etiology. Infection with high-risk human papillomavirus (hrHPV) has been shown to be a causative factor for cervical carcinogenesis. Despite the occurrence of prophylactic vaccines, highly sensitive HPV diagnostics, and innovative new therapies, cervical cancer remains a main cause of death in developing countries. The relationship between hrHPV infection and cervical cancer depends on the integration of viral DNA to the host genome, disrupting the viral regulator E2 and the continuous production of the viral E6 and E7 proteins, which are necessary to acquire and maintain a transformed phenotype but insufficient for malignant cervical carcinogenesis. Lately, miRNAs, the tumor microenvironment, and immune evasion have been found to be major players in cervical carcinogenesis after hrHPV infection. Many miRNAs have been widely reported as deregulated in cervical cancer. Here, the relevance of miRNA in HPV-mediated transformation is critically reviewed in the context of the natural history of hrHPV infection and cervical cancer.
Collapse
Affiliation(s)
- Giovanni Palomino-Vizcaino
- Facultad de Ciencias de la Salud, Unidad Valle de las Palmas, Campus Tijuana, Universidad Autónoma de Baja California, Tijuana 21500, Mexico;
| | - Evelyn Gabriela Bañuelos-Villegas
- Laboratorio de Terapia Génica, Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N., México City 07360, Mexico;
| | - Luis Marat Alvarez-Salas
- Laboratorio de Terapia Génica, Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del I.P.N., México City 07360, Mexico;
| |
Collapse
|
2
|
Nan F, Liu B, Yao C. Discovering the role of microRNAs and exosomal microRNAs in chest and pulmonary diseases: a spotlight on chronic obstructive pulmonary disease. Mol Genet Genomics 2024; 299:107. [PMID: 39527303 DOI: 10.1007/s00438-024-02199-2] [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: 09/20/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive respiratory condition and ranks as the fourth leading cause of mortality worldwide. Despite extensive research efforts, a reliable diagnostic or prognostic tool for COPD remains elusive. The identification of novel biomarkers may facilitate improved therapeutic strategies for patients suffering from this debilitating disease. MicroRNAs (miRNAs), which are small non-coding RNA molecules, have emerged as promising candidates for the prediction and diagnosis of COPD. Studies have demonstrated that dysregulation of miRNAs influences critical cellular and molecular pathways, including Notch, Wnt, hypoxia-inducible factor-1α, transforming growth factor, Kras, and Smad, which may contribute to the pathogenesis of COPD. Extracellular vesicles, particularly exosomes, merit further investigation due to their capacity to transport various biomolecules such as mRNAs, miRNAs, and proteins between cells. This intercellular communication can significantly impact the progression and severity of COPD by modulating signaling pathways in recipient cells. A deeper exploration of circulating miRNAs and the content of extracellular vesicles may lead to the discovery of novel diagnostic and prognostic biomarkers, ultimately enhancing the management of COPD. The current review focus on the pathogenic role of miRNAs and their exosomal counterparts in chest and respiratory diseases, centering COPD.
Collapse
Affiliation(s)
- FangYuan Nan
- Thoracic Surgery Department of the First People's Hospital of Jiangxia District, Wuhan, 430200, Hubei Province, China
| | - Bo Liu
- Thoracic Surgery Department of the First People's Hospital of Jiangxia District, Wuhan, 430200, Hubei Province, China
| | - Cheng Yao
- Infectious Diseases Department of the First People's Hospital of Jiangxia District, Wuhan, 430200, Hubei Province, China.
| |
Collapse
|
3
|
Wang D, Wang T, Kim D, Tan S, Liu S, Wan J, Deng Q. MicroRNA-375 modulates neutrophil chemotaxis via targeting Cathepsin B in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109933. [PMID: 39343064 PMCID: PMC11561466 DOI: 10.1016/j.fsi.2024.109933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/01/2024]
Abstract
Neutrophils are crucial for defense against numerous infections, and their migration and activations are tightly regulated to prevent collateral tissue damage. We previously performed a neutrophil-specific miRNA overexpression screening and identified several microRNAs, including miR-375, as potent modulators for neutrophil activity. Overexpression of miR-375 decreases neutrophil motility and migration in zebrafish and human neutrophil-like cells. We screened the genes downregulated by miR-375 in zebrafish neutrophils and identified that Cathepsin B (Ctsba) is required for neutrophil motility and chemotaxis upon tail wounding and bacterial infection. Pharmacological inhibition or neutrophil-specific knockout of ctsba significantly decreased the neutrophil chemotaxis in zebrafish and survival upon systemic bacterial infection. Notably, Ctsba knockdown in human neutrophil-like cells also resulted in reduced chemotaxis. Inhibiting integrin receptor function using RGDS rescued the neutrophil migration defects and susceptibility to systemic infection in zebrafish with either miR-375 overexpression or ctsba knockout. Our results demonstrate that miR-375 and its target Ctsba modulate neutrophil activity during tissue injury and bacterial infection in vivo, providing novel insights into neutrophil biology and the overall inflammation process.
Collapse
Affiliation(s)
- Decheng Wang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Tianqi Wang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Daniel Kim
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Shelly Tan
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Collaborative Core for Cancer Bioinformatics, Indiana University Simon Cancer Center, Indianapolis, IN, 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Collaborative Core for Cancer Bioinformatics, Indiana University Simon Cancer Center, Indianapolis, IN, 46202, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Qing Deng
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
4
|
Thawanaphong S, Nair A, Volfson E, Nair P, Mukherjee M. IL-18 biology in severe asthma. Front Med (Lausanne) 2024; 11:1486780. [PMID: 39554494 PMCID: PMC11566457 DOI: 10.3389/fmed.2024.1486780] [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: 08/26/2024] [Accepted: 10/18/2024] [Indexed: 11/19/2024] Open
Abstract
The role of interleukin-18 (IL-18) and inflammasomes in chronic inflammatory airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD), has garnered significant attention in recent years. This review aims to provide an overview of the current understanding of IL-18 biology, the associated signaling pathways, and the involvement of inflammasome complexes in airway diseases. We explore the multifaceted role of IL-18 in asthma pathophysiology, including its interactions with other cytokines and contributions to both T2 and non-T2 inflammation. Importantly, emerging evidence highlights IL-18 as a critical player in severe asthma, contributing to chronic airway inflammation, airway hyperresponsiveness (AHR), and mucus impaction. Furthermore, we discuss the emerging evidence of IL-18's involvement in autoimmunity and highlight potential therapeutic targets within the IL-18 and inflammasome pathways in severe asthma patients with evidence of infections and airway autoimmune responses. By synthesizing recent advancements and ongoing research, this review underscores the importance of IL-18 as a potential novel therapeutic target in the treatment of severe asthma and other related conditions.
Collapse
Affiliation(s)
- Sarita Thawanaphong
- Department of Medicine, McMAster University, Hamilton, ON, Canada
- Research Institute of St. Joe’s Hamilton, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Aswathi Nair
- Department of Medicine, McMAster University, Hamilton, ON, Canada
- Research Institute of St. Joe’s Hamilton, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
| | - Emily Volfson
- Department of Medicine, McMAster University, Hamilton, ON, Canada
- Research Institute of St. Joe’s Hamilton, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
| | - Parameswaran Nair
- Department of Medicine, McMAster University, Hamilton, ON, Canada
- Research Institute of St. Joe’s Hamilton, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
| | - Manali Mukherjee
- Department of Medicine, McMAster University, Hamilton, ON, Canada
- Research Institute of St. Joe’s Hamilton, St. Joseph’s Healthcare Hamilton, Hamilton, ON, Canada
| |
Collapse
|
5
|
Sandoval C, Nisson K, Fregoso OI. HIV-1 Vpr-induced DNA damage activates NF-κB through ATM-NEMO independent of cell cycle arrest. mBio 2024; 15:e0024024. [PMID: 39269169 PMCID: PMC11481869 DOI: 10.1128/mbio.00240-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 08/01/2024] [Indexed: 09/15/2024] Open
Abstract
Lentiviruses encode a number of multi-functional accessory proteins, however, the primary role of the accessory protein Vpr remains unclear. As Vpr engages the host DNA damage response (DDR) at multiple steps, modulation of the DDR is considered central to the function(s) of Vpr. Vpr activates ataxia telangiectasia and Rad3 (ATR)-mediated DDR signaling, resulting in cell cycle arrest. However, the cellular consequences of Vpr-induced DNA damage, and the connection of Vpr-induced DNA damage to other Vpr functions, are unknown. Here, we determined that HIV-1 Vpr-induced DNA damage activates the ATM-NF-κB essential modulator (NEMO) pathway and alters cellular transcription via NF-κB/RelA. Through RNA-sequencing (RNA-seq) of cells expressing Vpr or mutants that separate the ability of Vpr to induce DNA damage from other DDR phenotypes, we identified that Vpr alters the transcriptome independent of cell cycle arrest. In tissue-cultured U2OS cells and primary human monocyte-derived macrophages (MDMs), we showed Vpr activates both ataxia telangiectasia mutated (ATM) and NF-κB/RelA signaling cascades. While inhibition of NEMO did not affect Vpr-induced DNA damage, it prevented NF-κB activation by Vpr, highlighting the importance of NEMO in Vpr-mediated transcriptional reprogramming. Virion-delivered Vpr was sufficient to induce DNA damage and activate ATM-NEMO dependent NF-κB transcription, suggesting that engagement of the DDR and transcriptional changes can occur early during viral replication. Together, our data uncover cellular consequences of Vpr-induced DNA damage and provide a mechanism for how Vpr activates NF-κB through DNA damage and ATM-NEMO signaling, which occur independent of cell cycle arrest. We propose this is essential to overcoming restrictive environments, such as in macrophages, to enhance viral replication.IMPORTANCEThe HIV accessory protein Vpr is multi-functional and required for viral replication in vivo, yet how Vpr enhances viral replication is unknown. Emerging literature suggests that a conserved function of Vpr is the engagement of the host DNA damage response (DDR). For example, Vpr activates DDR signaling, causes DDR-dependent cell cycle arrest, promotes degradation of various DDR proteins, and alters cellular consequences of DDR activation. However, a central understanding of how these phenotypes connect and how they affect HIV-infected cells remains unknown. Here, we found that Vpr-induced DNA damage alters the host transcriptome by activating an essential transcription pathway, NF-κB. This occurs early during the infection of primary human immune cells, suggesting NF-κB activation and transcriptome remodeling are important for establishing productive HIV-1 infection. Together, our study provides novel insights into how Vpr alters the host environment through the DDR, and what roles Vpr and the DDR play to enhance HIV replication.
Collapse
Affiliation(s)
- Carina Sandoval
- Molecular Biology Institute, University of California, Los Angeles, California, USA
| | - Karly Nisson
- Molecular Biology Institute, University of California, Los Angeles, California, USA
| | - Oliver I. Fregoso
- Molecular Biology Institute, University of California, Los Angeles, California, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA
| |
Collapse
|
6
|
Mesaros O, Veres S, Onciul M, Matei E, Jimbu L, Neaga A, Zdrenghea M. Dysregulated MicroRNAs in Chronic Lymphocytic Leukemia. Cureus 2024; 16:e68770. [PMID: 39376808 PMCID: PMC11456419 DOI: 10.7759/cureus.68770] [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] [Accepted: 09/06/2024] [Indexed: 10/09/2024] Open
Abstract
MiRNAs are a class of non-coding RNAs acting as gene expression regulators by modulating the lifespan of messenger RNA. Commonly referred to as the most frequent leukemia in the Western world, chronic lymphocytic leukemia (CLL) is a lymphoproliferative malignancy characterized by clonal expansion of CD19, CD23, and CD5-positive mature B-cells. While this pathology is regarded as less aggressive and has a variety of treatment options, the cause of its clinical heterogeneity is not yet understood. Moreover, the prognostic markers and treatment recommendations based on predictive markers are limited. This review aims to investigate some miRNAs that are dysregulated and possibly involved in CLL pathogenesis as a starting point for the proposal of new prognostic and predictive markers and, as more agents targeting miRNA expression become available, their potential role as therapeutic targets.
Collapse
Affiliation(s)
- Oana Mesaros
- Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, ROU
- Hematology, Ion Chiricuta Oncology Institute, Cluj-Napoca, ROU
| | - Stefana Veres
- Otolaryngology, Policlinica Grigorescu, Cluj-Napoca, ROU
| | - Madalina Onciul
- Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, ROU
| | - Emilia Matei
- Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, ROU
- Pathology, Ion Chiricuta Oncology Institute, Cluj-Napoca, ROU
| | - Laura Jimbu
- Hematology, Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, ROU
- Hematology, Ion Chiricuta Oncology Institute, Cluj-Napoca, ROU
| | - Alexandra Neaga
- Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, ROU
| | - Mihnea Zdrenghea
- Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, ROU
| |
Collapse
|
7
|
Ma H, Ou ZL, Alaeiilkhchi N, Cheng YQ, Chen K, Chen JY, Guo RQ, He MY, Tang SY, Zhang X, Huang ZP, Liu J, Liu J, Zhu QA, Huang ZC, Jiang H. MiR-223 enhances lipophagy by suppressing CTSB in microglia following lysolecithin-induced demyelination in mice. Lipids Health Dis 2024; 23:194. [PMID: 38909243 PMCID: PMC11193262 DOI: 10.1186/s12944-024-02185-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Lipid droplet (LD)-laden microglia is a key pathological hallmark of multiple sclerosis. The recent discovery of this novel microglial subtype, lipid-droplet-accumulating microglia (LDAM), is notable for increased inflammatory factor secretion and diminished phagocytic capability. Lipophagy, the autophagy-mediated selective degradation of LDs, plays a critical role in this context. This study investigated the involvement of microRNAs (miRNAs) in lipophagy during demyelinating diseases, assessed their capacity to modulate LDAM subtypes, and elucidated the potential underlying mechanisms involved. METHODS C57BL/6 mice were used for in vivo experiments. Two weeks post demyelination induction at cervical level 4 (C4), histological assessments and confocal imaging were performed to examine LD accumulation in microglia within the lesion site. Autophagic changes were observed using transmission electron microscopy. miRNA and mRNA multi-omics analyses identified differentially expressed miRNAs and mRNAs under demyelinating conditions and the related autophagy target genes. The role of miR-223 in lipophagy under these conditions was specifically explored. In vitro studies, including miR-223 upregulation in BV2 cells via lentiviral infection, validated the bioinformatics findings. Immunofluorescence staining was used to measure LD accumulation, autophagy levels, target gene expression, and inflammatory mediator levels to elucidate the mechanisms of action of miR-223 in LDAM. RESULTS Oil Red O staining and confocal imaging revealed substantial LD accumulation in the demyelinated spinal cord. Transmission electron microscopy revealed increased numbers of autophagic vacuoles at the injury site. Multi-omics analysis revealed miR-223 as a crucial regulatory gene in lipophagy during demyelination. It was identified that cathepsin B (CTSB) targets miR-223 in autophagy to integrate miRNA, mRNA, and autophagy gene databases. In vitro, miR-223 upregulation suppressed CTSB expression in BV2 cells, augmented autophagy, alleviated LD accumulation, and decreased the expression of the inflammatory mediator IL-1β. CONCLUSION These findings indicate that miR-223 plays a pivotal role in lipophagy under demyelinating conditions. By inhibiting CTSB, miR-223 promotes selective LD degradation, thereby reducing the lipid burden and inflammatory phenotype in LDAM. This study broadens the understanding of the molecular mechanisms of lipophagy and proposes lipophagy induction as a potential therapeutic approach to mitigate inflammatory responses in demyelinating diseases.
Collapse
Grants
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No. 81972064 National Natural Science Foundation of China
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2020A1515011415 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- 202102021244 Guangzhou Science and Technology Plan Project
Collapse
Affiliation(s)
- Hao Ma
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhi-Lin Ou
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Nima Alaeiilkhchi
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia, Vancouver, Canada
| | - Yong-Quan Cheng
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Kai Chen
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jia-Yu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Ru-Qin Guo
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Min-Yue He
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Shi-Yi Tang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xin Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhi-Ping Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Junhao Liu
- Division of Spine Surgery, Department of Orthopaedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 51000, China
| | - Jie Liu
- The 3rd Affiliated Hospital of Xinxiang Medical University, Henan, China
| | - Qing-An Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zu-Cheng Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Hui Jiang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
8
|
Gordon S, Layton AM, Fawcett S, Ross K. A microRNA focus on acne. Dermatol Reports 2024; 16:9902. [PMID: 38957637 PMCID: PMC11216150 DOI: 10.4081/dr.2024.9902] [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: 11/26/2023] [Accepted: 12/03/2023] [Indexed: 07/04/2024] Open
Abstract
Acne (syn. acne vulgaris) is a common inflammatory skin disorder associated with puberty and adolescence. The disease is characterized by comedoneous lesions, papules, pustules, and nodules that are mostly found on the face. These lesions are caused by intricate interactions between the pilosebaceous unit and the Cutibacterium acnes (C. acnes) bacteria. Unhealthy acne and its aftereffects, like pigment changes and scarring, have a detrimental impact on one's quality of life. Recent years have seen a sharp increase in the approval of nucleic acid therapies (NATs), such as antisense oligonucleotides and short-interfering RNA medications, for rare diseases for which there are few or no effective treatments. These developments suggest that NATs may be useful in acne treatment plans down the road, as do clinical trials for microRNA (miRNA) modulation in skin contexts. We highlight promising miRNA targets for anti-acne therapy in this review. We outline the pathophysiology of acne in brief and emphasize the functions of C. acnes. Next, we concentrate on the distinct impacts of biofilm and planktonic C. acnes on a Toll-like receptor 2 axis that spans miR-146a-5p, which was recently discovered. Before discussing the potential contributions of miR-21-5p, miR-233-3p, and miR-150-5p to inflammatory axes in acne, we evaluate miR-146a-5p in sebocytes. Finally, we address patient involvement in miRNA-related acne research and translational perspectives.
Collapse
Affiliation(s)
| | - Alison M. Layton
- Skin Research Centre, Hull York Medical School, University of York
- Harrogate and District NHS Foundation Trust, Harrogate
| | - Sandra Fawcett
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University
- Institute for Health Research, Liverpool John Moores University, United Kingdom
| | - Kehinde Ross
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University
- Institute for Health Research, Liverpool John Moores University, United Kingdom
| |
Collapse
|
9
|
Cihankaya H, Bader V, Winklhofer KF, Vorgerd M, Matschke J, Stahlke S, Theiss C, Matschke V. Elevated NLRP3 Inflammasome Activation Is Associated with Motor Neuron Degeneration in ALS. Cells 2024; 13:995. [PMID: 38920626 PMCID: PMC11202041 DOI: 10.3390/cells13120995] [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: 04/25/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by motor neuron degeneration in the central nervous system. Recent research has increasingly linked the activation of nucleotide oligomerization domain-like receptor protein 3 (NLRP3) inflammasome to ALS pathogenesis. NLRP3 activation triggers Caspase 1 (CASP 1) auto-activation, leading to the cleavage of Gasdermin D (GSDMD) and pore formation on the cellular membrane. This process facilitates cytokine secretion and ultimately results in pyroptotic cell death, highlighting the complex interplay of inflammation and neurodegeneration in ALS. This study aimed to characterize the NLRP3 inflammasome components and their colocalization with cellular markers using the wobbler mouse as an ALS animal model. Firstly, we checked the levels of miR-223-3p because of its association with NLRP3 inflammasome activity. The wobbler mice showed an increased expression of miR-223-3p in the ventral horn, spinal cord, and cerebellum tissues. Next, increased levels of NLRP3, pro-CASP 1, cleaved CASP 1 (c-CASP 1), full-length GSDMD, and cleaved GDSMD revealed NLRP3 inflammasome activation in wobbler spinal cords, but not in the cerebellum. Furthermore, we investigated the colocalization of the aforementioned proteins with neurons, microglia, and astrocyte markers in the spinal cord tissue. Evidently, the wobbler mice displayed microgliosis, astrogliosis, and motor neuron degeneration in this tissue. Additionally, we showed the upregulation of protein levels and the colocalization of NLRP3, c-CASP1, and GSDMD in neurons, as well as in microglia and astrocytes. Overall, this study demonstrated the involvement of NLRP3 inflammasome activation and pyroptotic cell death in the spinal cord tissue of wobbler mice, which could further exacerbate the motor neuron degeneration and neuroinflammation in this ALS mouse model.
Collapse
Affiliation(s)
- Hilal Cihankaya
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, 44801 Bochum, Germany; (H.C.); (C.T.)
- International Graduate School of Neuroscience (IGSN), Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Verian Bader
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Konstanze F. Winklhofer
- International Graduate School of Neuroscience (IGSN), Ruhr-University Bochum, 44801 Bochum, Germany;
- Department of Molecular Cell Biology, Institute of Biochemistry and Pathobiochemistry, Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Buerkle-de-la-Camp-Platz 1, 44789 Bochum, Germany;
| | - Johann Matschke
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Sarah Stahlke
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, 44801 Bochum, Germany; (H.C.); (C.T.)
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, 44801 Bochum, Germany; (H.C.); (C.T.)
- International Graduate School of Neuroscience (IGSN), Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, 44801 Bochum, Germany; (H.C.); (C.T.)
- International Graduate School of Neuroscience (IGSN), Ruhr-University Bochum, 44801 Bochum, Germany;
| |
Collapse
|
10
|
Li MY, Chong LC, Duns G, Lytle A, Woolcock B, Jiang A, Telenius A, Ben-Neriah S, Nawaz W, Slack GW, Elisia I, Viganò E, Aoki T, Healy S, Krystal G, Venturutti L, Scott DW, Steidl C. TRAF3 loss-of-function reveals the noncanonical NF-κB pathway as a therapeutic target in diffuse large B cell lymphoma. Proc Natl Acad Sci U S A 2024; 121:e2320421121. [PMID: 38662551 PMCID: PMC11067025 DOI: 10.1073/pnas.2320421121] [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: 11/20/2023] [Accepted: 03/29/2024] [Indexed: 05/05/2024] Open
Abstract
Here, we report recurrent focal deletions of the chr14q32.31-32 locus, including TRAF3, a negative regulator of NF-κB signaling, in de novo diffuse large B cell lymphoma (DLBCL) (24/324 cases). Integrative analysis revealed an association between TRAF3 copy number loss with accumulation of NIK, the central noncanonical (NC) NF-κB kinase, and increased NC NF-κB pathway activity. Accordingly, TRAF3 genetic ablation in isogenic DLBCL model systems caused upregulation of NIK and enhanced NC NF-κB downstream signaling. Knockdown or pharmacological inhibition of NIK in TRAF3-deficient cells differentially impaired their proliferation and survival, suggesting an acquired onco-addiction to NC NF-κB. TRAF3 ablation also led to exacerbated secretion of the immunosuppressive cytokine IL-10. Coculturing of TRAF3-deficient DLBCL cells with CD8+ T cells impaired the induction of Granzyme B and interferon (IFN) γ, which were restored following neutralization of IL-10. Our findings corroborate a direct relationship between TRAF3 genetic alterations and NC NF-κB activation, and highlight NIK as a potential therapeutic target in a defined subset of DLBCL.
Collapse
Affiliation(s)
- Michael Y. Li
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
| | - Lauren C. Chong
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Gerben Duns
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Andrew Lytle
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Bruce Woolcock
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Aixiang Jiang
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
| | - Adèle Telenius
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Susana Ben-Neriah
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Waqas Nawaz
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Graham W. Slack
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
| | - Ingrid Elisia
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Elena Viganò
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Tomohiro Aoki
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Shannon Healy
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Gerald Krystal
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Leandro Venturutti
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - David W. Scott
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
| | - Christian Steidl
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
| |
Collapse
|
11
|
Hanley S, Chen YY, Hazeldine J, Lord JM. Senescent cell-derived extracellular vesicles as potential mediators of innate immunosenescence and inflammaging. Exp Gerontol 2024; 187:112365. [PMID: 38237747 DOI: 10.1016/j.exger.2024.112365] [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: 12/01/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/27/2024]
Abstract
Ageing is accompanied by a decline in immune function (immunosenescence), increased inflammation (inflammaging), and more senescent cells which together contribute to age-related disease and infection susceptibility. The innate immune system is the front-line defence against infection and cancer and is also involved in the removal of senescent cells, so preventing innate immunosenescence and inflammaging is vital for health in older age. Extracellular vesicles (EVs) modulate many aspects of innate immune function, including chemotaxis, anti-microbial responses, and immune regulation. Senescent cell derived EVs (SEVs) have different cargo to that of non-senescent cell derived EVs, suggesting alterations in EV cargo across the lifespan may influence innate immune function, possibly contributing to immunosenescence and inflammaging. Here we review current understanding of the potential impact of miRNAs, lipids and proteins, found in higher concentrations in SEVs, on innate immune functions and inflammation to consider whether SEVs are potential influencers of innate immunosenescence and inflammaging. Furthermore, senolytics have demonstrated an ability to return plasma EV content closer to that of non-senescent EVs, therefore the potential use of senotherapeutics (senolytics and senostatics) to ameliorate the effects of SEVs on immunosenescence and inflammaging is also considered as a possible strategy for extending health-span in older adults.
Collapse
Affiliation(s)
- Shaun Hanley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Yung-Yi Chen
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Jon Hazeldine
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK
| | - Janet M Lord
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
12
|
Hattori T, Fundora KA, Hamamoto K, Opozda DM, Liang X, Liu X, Zhang J, Uzun Y, Takahashi Y, Wang HG. ER stress elicits non-canonical CASP8 (caspase 8) activation on autophagosomal membranes to induce apoptosis. Autophagy 2024; 20:349-364. [PMID: 37733908 PMCID: PMC10813646 DOI: 10.1080/15548627.2023.2258701] [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: 02/06/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
The VPS37A gene encodes a subunit of the endosomal sorting complex required for transport (ESCRT)-I complex that is frequently lost in a wide variety of human solid cancers. We have previously demonstrated the role of VPS37A in directing the ESCRT membrane scission machinery to seal the phagophore for autophagosome completion. Here, we report that VPS37A-deficient cells exhibit an accumulation of the apoptotic initiator CASP8 (caspase 8) on the phagophore and are primed to undergo rapid apoptosis through the intracellular death-inducing signaling complex (iDISC)-mediated CASP8 activation upon exposure to endoplasmic reticulum (ER) stress. Using CRISPR-Cas9 gene editing and comparative transcriptome analysis, we identified the ATF4-mediated stress response pathway as a crucial mediator to elicit iDISC-mediated apoptosis following the inhibition of autophagosome closure. Notably, ATF4-mediated iDISC activation occurred independently of the death receptor TNFRSF10B/DR5 upregulation but required the pro-apoptotic transcriptional factor DDIT3/CHOP to enhance the mitochondrial amplification pathway for full-activation of CASP8 in VPS37A-deficient cells stimulated with ER stress inducers. Our analysis also revealed the upregulation of NFKB/NF-kB signaling as a potential mechanism responsible for restraining iDISC activation and promoting cell survival upon VPS37A depletion. These findings have important implications for the future development of new strategies to treat human cancers, especially those with VPS37A loss.Abbreviations: ATG: autophagy related; BMS: BMS-345541; CASP: caspase; CHMP: charged multivesicular body protein; DKO: double knockout; Dox: doxycycline; ER: endoplasmic reticulum; ESCRT: endosomal sorting complex required for transport; gRNA: guide RNA; GSEA: gene set enrichment analysis; GSK157: GSK2656157; iDISC: intracellular death-inducing signaling complex; IKK: inhibitor of NFKB kinase; IPA: ingenuity pathway analysis; KO: knockout; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; NFKB/NF-kB: nuclear factor kappa B; OZ: 5Z-7-oxozeaenol; RNA-seq: RNA sequencing; UPR: unfolded protein response; TFT: transcription factor target; THG: thapsigargin; TUN: tunicamycin; VPS: vacuolar protein sorting.
Collapse
Affiliation(s)
- Tatsuya Hattori
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Kevin A. Fundora
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Kouta Hamamoto
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - David M. Opozda
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Xinwen Liang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Xiaoming Liu
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Jiawen Zhang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Yasin Uzun
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Yoshinori Takahashi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Hong-Gang Wang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
13
|
Shi M, Lu Q, Zhao Y, Ding Z, Yu S, Li J, Ji M, Fan H, Hou S. miR-223: a key regulator of pulmonary inflammation. Front Med (Lausanne) 2023; 10:1187557. [PMID: 37465640 PMCID: PMC10350674 DOI: 10.3389/fmed.2023.1187557] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/14/2023] [Indexed: 07/20/2023] Open
Abstract
Small noncoding RNAs, known as microRNAs (miRNAs), are vital for the regulation of diverse biological processes. miR-223, an evolutionarily conserved anti-inflammatory miRNA expressed in cells of the myeloid lineage, has been implicated in the regulation of monocyte-macrophage differentiation, proinflammatory responses, and the recruitment of neutrophils. The biological functions of this gene are regulated by its expression levels in cells or tissues. In this review, we first outline the regulatory role of miR-223 in granulocytes, macrophages, endothelial cells, epithelial cells and dendritic cells (DCs). Then, we summarize the possible role of miR-223 in chronic obstructive pulmonary disease (COPD), acute lung injury (ALI), coronavirus disease 2019 (COVID-19) and other pulmonary inflammatory diseases to better understand the molecular regulatory networks in pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Mingyu Shi
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Ziling Ding
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Sifan Yu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Junfeng Li
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Mengjun Ji
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
| |
Collapse
|
14
|
Dancik GM, Varisli L, Vlahopoulos SA. The Molecular Context of Oxidant Stress Response in Cancer Establishes ALDH1A1 as a Critical Target: What This Means for Acute Myeloid Leukemia. Int J Mol Sci 2023; 24:ijms24119372. [PMID: 37298333 DOI: 10.3390/ijms24119372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The protein family of aldehyde dehydrogenases (ALDH) encompasses nineteen members. The ALDH1 subfamily consists of enzymes with similar activity, having the capacity to neutralize lipid peroxidation products and to generate retinoic acid; however, only ALDH1A1 emerges as a significant risk factor in acute myeloid leukemia. Not only is the gene ALDH1A1 on average significantly overexpressed in the poor prognosis group at the RNA level, but its protein product, ALDH1A1 protects acute myeloid leukemia cells from lipid peroxidation byproducts. This capacity to protect cells can be ascribed to the stability of the enzyme under conditions of oxidant stress. The capacity to protect cells is evident both in vitro, as well as in mouse xenografts of those cells, shielding cells effectively from a number of potent antineoplastic agents. However, the role of ALDH1A1 in acute myeloid leukemia has been unclear in the past due to evidence that normal cells often have higher aldehyde dehydrogenase activity than leukemic cells. This being true, ALDH1A1 RNA expression is significantly associated with poor prognosis. It is hence imperative that ALDH1A1 is methodically targeted, particularly for the acute myeloid leukemia patients of the poor prognosis risk group that overexpress ALDH1A1 RNA.
Collapse
Affiliation(s)
- Garrett M Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA
| | - Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey
| | - Spiros A Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527 Athens, Greece
| |
Collapse
|
15
|
Monda JK, Ge X, Hunkeler M, Donovan KA, Ma MW, Jin CY, Leonard M, Fischer ES, Bennett EJ. HAPSTR1 localizes HUWE1 to the nucleus to limit stress signaling pathways. Cell Rep 2023; 42:112496. [PMID: 37167062 DOI: 10.1016/j.celrep.2023.112496] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/21/2023] [Accepted: 04/25/2023] [Indexed: 05/13/2023] Open
Abstract
HUWE1 is a large, enigmatic HECT-domain ubiquitin ligase implicated in the regulation of diverse pathways, including DNA repair, apoptosis, and differentiation. How HUWE1 engages its structurally diverse substrates and how HUWE1 activity is regulated are unknown. Using unbiased quantitative proteomics, we find that HUWE1 targets substrates in a largely cell-type-specific manner. However, we identify C16orf72/HAPSTR1 as a robust HUWE1 substrate in multiple cell lines. Previously established physical and genetic interactions between HUWE1 and HAPSTR1 suggest that HAPSTR1 positively regulates HUWE1 function. Here, we show that HAPSTR1 is required for HUWE1 nuclear localization and nuclear substrate targeting. Nuclear HUWE1 is required for both cell proliferation and modulation of stress signaling pathways, including p53 and nuclear factor κB (NF-κB)-mediated signaling. Combined, our results define a role for HAPSTR1 in gating critical nuclear HUWE1 functions.
Collapse
Affiliation(s)
- Julie K Monda
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xuezhen Ge
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Moritz Hunkeler
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine A Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Michelle W Ma
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Cyrus Y Jin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Marilyn Leonard
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Eric J Bennett
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
16
|
Li H, Lv J, Wang J, Wang H, Luo L. MiR-29c-3p represses gastric cancer development via modulating MEST. Histol Histopathol 2023; 38:549-557. [PMID: 36269039 DOI: 10.14670/hh-18-537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Gastric cancer (GC) triggers a great number of deaths worldwide. Although great efforts have been made in treating this cancer, GC patients' survival rate remains unsatisfactory. An increasing amount of evidence indicates that miR-29c-3p inhibits cancer progression. However, the mechanism of miR-29c-3p in GC remains to be fully defined. Hence, this work aimed to analyze the underlying mechanism of miR-29c-3p in GC. Outcomes showed marked downregulation of miR-29c-3p in GC tissue and cell lines. Functional experiments exhibited that miR-29c-3p repressed GC cell malignant behaviors. Moreover, bioinformatics analysis and dual-luciferase reporter gene detection indicated that MEST was targeted by miR-29c-3p. Rescue assay further proved that MEST participated in functions of miR-29c-3p in GC. To sum up, miR-29c-3p/MEST signaling pathway suppressed formation of malignant phenotypes of GC, and targeting the signaling pathway may be a new method for treating GC.
Collapse
Affiliation(s)
- Honghai Li
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing City, Zhejiang Province, PR China
| | - Jieqing Lv
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing City, Zhejiang Province, PR China
| | - Jindao Wang
- Department of Endoscopy Center, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing City, Zhejiang Province, PR China
| | - Haifeng Wang
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing City, Zhejiang Province, PR China
| | - Liang Luo
- Department of Radiotherapy, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing City, Zhejiang Province, PR China.
| |
Collapse
|
17
|
Goekeri C, Pennitz P, Groenewald W, Behrendt U, Kirsten H, Zobel CM, Berger S, Heinz GA, Mashreghi MF, Wienhold SM, Dietert K, Dorhoi A, Gruber AD, Scholz M, Rohde G, Suttorp N, Witzenrath M, Nouailles G. MicroRNA-223 Dampens Pulmonary Inflammation during Pneumococcal Pneumonia. Cells 2023; 12:cells12060959. [PMID: 36980300 PMCID: PMC10047070 DOI: 10.3390/cells12060959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
Community-acquired pneumonia remains a major contributor to global communicable disease-mediated mortality. Neutrophils play a leading role in trying to contain bacterial lung infection, but they also drive detrimental pulmonary inflammation, when dysregulated. Here we aimed at understanding the role of microRNA-223 in orchestrating pulmonary inflammation during pneumococcal pneumonia. Serum microRNA-223 was measured in patients with pneumococcal pneumonia and in healthy subjects. Pulmonary inflammation in wild-type and microRNA-223-knockout mice was assessed in terms of disease course, histopathology, cellular recruitment and evaluation of inflammatory protein and gene signatures following pneumococcal infection. Low levels of serum microRNA-223 correlated with increased disease severity in pneumococcal pneumonia patients. Prolonged neutrophilic influx into the lungs and alveolar spaces was detected in pneumococci-infected microRNA-223-knockout mice, possibly accounting for aggravated histopathology and acute lung injury. Expression of microRNA-223 in wild-type mice was induced by pneumococcal infection in a time-dependent manner in whole lungs and lung neutrophils. Single-cell transcriptome analyses of murine lungs revealed a unique profile of antimicrobial and cellular maturation genes that are dysregulated in neutrophils lacking microRNA-223. Taken together, low levels of microRNA-223 in human pneumonia patient serum were associated with increased disease severity, whilst its absence provoked dysregulation of the neutrophil transcriptome in murine pneumococcal pneumonia.
Collapse
Affiliation(s)
- Cengiz Goekeri
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Faculty of Medicine, Cyprus International University, 99040 Nicosia, Cyprus
- Correspondence: (C.G.); (G.N.)
| | - Peter Pennitz
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Wibke Groenewald
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Ulrike Behrendt
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics, and Epidemiology, Universität Leipzig, 04107 Leipzig, Germany
| | - Christian M. Zobel
- Department of Internal Medicine, Bundeswehrkrankenhaus Berlin, 10115 Berlin, Germany
| | - Sarah Berger
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Gitta A. Heinz
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), ein Institut der Leibniz-Gemeinschaft, 10117 Berlin, Germany
| | - Mir-Farzin Mashreghi
- Therapeutic Gene Regulation, Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), ein Institut der Leibniz-Gemeinschaft, 10117 Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany
| | - Sandra-Maria Wienhold
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Kristina Dietert
- Institute of Veterinary Pathology, Freie Universität Berlin, 14163 Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, 14163 Berlin, Germany
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, 17489 Greifswald, Germany
| | - Achim D. Gruber
- Institute of Veterinary Pathology, Freie Universität Berlin, 14163 Berlin, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics, and Epidemiology, Universität Leipzig, 04107 Leipzig, Germany
| | - Gernot Rohde
- Department of Respiratory Medicine, Medical Clinic I, Goethe-Universität Frankfurt am Main, 60596 Frankfurt am Main, Germany
- CAPNETZ STIFTUNG, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625 Hannover, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- CAPNETZ STIFTUNG, 30625 Hannover, Germany
- German Center for Lung Research (DZL), 10117 Berlin, Germany
| | | | - Martin Witzenrath
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- CAPNETZ STIFTUNG, 30625 Hannover, Germany
- German Center for Lung Research (DZL), 10117 Berlin, Germany
| | - Geraldine Nouailles
- Department of Infectious Diseases, Respiratory Medicine and Critical Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Correspondence: (C.G.); (G.N.)
| |
Collapse
|
18
|
Sullivan T, Sharma A, Lamers K, Mallard BA, Cánovas A, Karrow NA. Immunotolerance of dairy heifers in response to repeated exposure to bacterial lipopolysaccharide endotoxin. Vet Immunol Immunopathol 2023; 259:110579. [PMID: 36948027 DOI: 10.1016/j.vetimm.2023.110579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/30/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
Dairy cattle face a variety of stressful events on a daily basis. More specifically, climate change has resulted in more frequent heat stress events that increase the incidence of chronic bacterial infections by inducing conditions like leaky gut syndrome, whereby the integrity of the intestinal epithelium is compromised allowing for luminal bacterial lipopolysaccharide (LPS) endotoxin to infiltrate the host's bloodstream resulting in acute or chronic systemic stimulation of the innate immune system. Repeated exposure to LPS over a short period of time is reported to induce immunotolerance within the host. This LPS tolerance is an essential immunohomeostatic response that can protect against over activation of the inflammatory response during subsequent exposure to LPS. In the present study, Holstein calves (n = 20) were initially stress challenged with either saline, or 100, 200 or 400 ng/kg of LPS administered intramuscular, and again re-challenged with 200 ng/kg of LPS 2-weeks later. Serum was collected every 2 hr for 6 hr to profile changes in circulatory stress biomarkers after the repeated LPS exposures. Heifers that were initially challenged with 100, 200 and 400 ng/kg of LPS demonstrated significantly attenuated cortisol responses in the second challenge (p < 0.01, 0.01 and 0.05, respectively), whereas control animals who previously received saline demonstrated a strong cortisol response at 2 hr after receiving 200 ng/kg of LPS (p < 0.05). The cytokine/chemokine (IL-6, CCL2, CCL3 and CCL4) responses were also attenuated during the LPS rechallenge (p < 0.05). Finally, microRNA expression profiles were determined to assess the epigenetic response to repeated LPS exposure. Interestingly, miR-31 and miR-223 were downregulated in response to the second LPS challenge. The present study demonstrates the dynamic nature of the stress response in dairy cattle as it relates to the development of LPS tolerance. Understanding the roles of various stress biomarkers in the context of innate immune cell tolerance is essential for evaluating their impact on immune system homeostasis.
Collapse
Affiliation(s)
- T Sullivan
- Department of Animal Biosciences, Center for Genetic Improvement of Livestock, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - A Sharma
- Department of Animal Biosciences, Center for Genetic Improvement of Livestock, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - K Lamers
- Department of Animal Biosciences, Center for Genetic Improvement of Livestock, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - B A Mallard
- Department of Pathobiology, Ontario Veterinary College, Guelph, ON N1G 2W1, Canada
| | - A Cánovas
- Department of Animal Biosciences, Center for Genetic Improvement of Livestock, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - N A Karrow
- Department of Animal Biosciences, Center for Genetic Improvement of Livestock, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
19
|
Geiger F, Zeitlmayr S, Staab-Weijnitz CA, Rajan S, Breit A, Gudermann T, Dietrich A. An Inhibitory Function of TRPA1 Channels in TGF-β1-driven Fibroblast-to-Myofibroblast Differentiation. Am J Respir Cell Mol Biol 2023; 68:314-325. [PMID: 36378826 DOI: 10.1165/rcmb.2022-0159oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
TRPA1 (transient receptor potential ankyrin 1) is a nonselective Ca2+-permeable cation channel, which was originally cloned from human lung fibroblasts (HLFs). TRPA1-mediated Ca2+ entry is evoked by exposure to several chemicals, including allyl isothiocyanate (AITC), and a protective effect of TRPA1 activation in the development of cardiac fibrosis has been proposed. Yet the function of TRPA1 in TGF-β1 (transforming growth factor-β1)-driven fibroblast-to-myofibroblast differentiation and the development of pulmonary fibrosis remains elusive. TRPA1 expression and function were analyzed in cultured primary HLFs, and mRNA concentrations were significantly reduced after adding TGF-β1. Expression of genes encoding fibrosis markers (e.g., ACTA2, SERPINE1 [plasminogen activator inhibitor 1], FN1 [fibronectin], COL1A1 [type I collagen]) was increased after siRNA-mediated downregulation of TRPA1 mRNA in HLFs. Moreover, AITC-induced Ca2+ entry in HLFs was decreased after TGF-β1 treatment and by application of TRPA1 siRNAs, while AITC treatment alone did not reduce cell viability or enhance apoptosis. Most interestingly, AITC-induced TRPA1 activation augmented ERK1/2 (extracellular signal-regulated kinase 1/2) and SMAD2 linker phosphorylation, which might inhibit TGF-β-receptor signaling. Our results suggest an inhibitory function of TRPA1 channels in TGF-β1-driven fibroblast-to-myofibroblast differentiation. Therefore, activation of TRPA1 channels might be protective during the development of pulmonary fibrosis in patients.
Collapse
Affiliation(s)
- Fabienne Geiger
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research, Ludwig-Maximilians-University Munich, Munich, Germany, and
| | - Sarah Zeitlmayr
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research, Ludwig-Maximilians-University Munich, Munich, Germany, and
| | - Claudia A Staab-Weijnitz
- Comprehensive Pneumology Center with the CPC-M BioArchive and Institute of Lung Health and Immunity, Helmholtz Center Munich, Member of the German Center for Lung Research, Munich, Germany
| | - Suhasini Rajan
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research, Ludwig-Maximilians-University Munich, Munich, Germany, and
| | - Andreas Breit
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research, Ludwig-Maximilians-University Munich, Munich, Germany, and
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research, Ludwig-Maximilians-University Munich, Munich, Germany, and
| | - Alexander Dietrich
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research, Ludwig-Maximilians-University Munich, Munich, Germany, and
| |
Collapse
|
20
|
Cai D, Sun C, Murashita T, Que X, Chen SY. ADAR1 Non-Editing Function in Macrophage Activation and Abdominal Aortic Aneurysm. Circ Res 2023; 132:e78-e93. [PMID: 36688311 PMCID: PMC10316962 DOI: 10.1161/circresaha.122.321722] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023]
Abstract
BACKGROUND Macrophage activation plays a critical role in abdominal aortic aneurysm (AAA) development. However, molecular mechanisms controlling macrophage activation and vascular inflammation in AAA remain largely unknown. The objective of the study was to identify novel mechanisms underlying adenosine deaminase acting on RNA (ADAR1) function in macrophage activation and AAA formation. METHODS Aortic transplantation was conducted to determine the importance of nonvascular ADAR1 in AAA development/dissection. Ang II (Angiotensin II) infusion of ApoE-/- mouse model combined with macrophage-specific knockout of ADAR1 was used to study ADAR1 macrophage-specific role in AAA formation/dissection. The relevance of macrophage ADAR1 to human AAA was examined using human aneurysm specimens. Moreover, a novel humanized AAA model was established to test the role of human macrophages in aneurysm formation in human arteries. RESULTS Allograft transplantation of wild-type abdominal aortas to ADAR1+/- recipient mice significantly attenuated AAA formation, suggesting that nonvascular ADAR1 is essential for AAA development. ADAR1 deficiency in hematopoietic cells decreased the prevalence and severity of AAA while inhibited macrophage infiltration and aorta wall inflammation. ADAR1 deletion blocked the classic macrophage activation, diminished NF-κB (nuclear factor kappa B) signaling, and enhanced the expression of a number of anti-inflammatory microRNAs. Mechanistically, ADAR1 interacted with Drosha to promote its degradation, which attenuated Drosha-DGCR8 (DiGeorge syndrome critical region 8) interaction, and consequently inhibited pri- to pre-microRNA processing of microRNAs targeting IKKβ, resulting in an increased IKKβ (inhibitor of nuclear factor kappa-B) expression and enhanced NF-κB signaling. Significantly, ADAR1 was induced in macrophages and interacted with Drosha in human AAA lesions. Reconstitution of ADAR1-deficient, but not the wild type, human monocytes to immunodeficient mice blocked the aneurysm formation in transplanted human arteries. CONCLUSIONS Macrophage ADAR1 promotes aneurysm formation in both mouse and human arteries through a novel mechanism, that is, Drosha protein degradation, which inhibits the processing of microRNAs targeting NF-kB signaling and thus elicits macrophage-mediated vascular inflammation in AAA.
Collapse
Affiliation(s)
- Dunpeng Cai
- Departments of Surgery, University of Missouri School of Medicine, Columbia, MO
| | - Chenming Sun
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA
| | - Takashi Murashita
- Departments of Surgery, University of Missouri School of Medicine, Columbia, MO
| | - Xingyi Que
- Departments of Surgery, University of Missouri School of Medicine, Columbia, MO
| | - Shi-You Chen
- Departments of Surgery, University of Missouri School of Medicine, Columbia, MO
- Department of Medical Pharmacology & Physiology, University of Missouri School of Medicine, Columbia, MO
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA
| |
Collapse
|
21
|
Mao B, Guo W, Liu X, Cui S, Zhang Q, Zhao J, Tang X, Zhang H. Potential Probiotic Properties of Blautia producta Against Lipopolysaccharide-Induced Acute Liver Injury. Probiotics Antimicrob Proteins 2023; 15:785-796. [PMID: 36790661 DOI: 10.1007/s12602-023-10044-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2023] [Indexed: 02/16/2023]
Abstract
Blautia is a genus of anaerobic microbe extensively present in the intestine and feces of mammals. This study aims to investigate the influence of Blautia producta to prevent lipopolysaccharide (LPS)-induced acute liver injury (ALI) and elaborate on its hepatoprotective mechanisms. B. producta D4 and DSM2950 pretreatment decreased the activities of serum aspartate transferase (AST), and alanine transaminase (ALT) in mice with LPS treatment significantly decreased the levels of inflammatory tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) and increased the activities of antioxidative superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px). Compared with the model group, B. producta D4 and B. producta DSM2950 pretreatment slightly increased the levels of cecal propionic acid, isobutyric acid, butyric acid, valeric acid, and isovaleric acid (p > 0.05). Metagenomic analysis showed that B. producta D4 and DSM2950 pretreatment remarkably increased the relative abundance of [Eubacterium] xylanophilum group, Lachnospira, Ruminiclostridium, Ruminiclostridium 9, Coprococcus 2, Odoribacter, Roseburia, Alistipes, and Desulfovibrio in ALI mice, and their abundance is negatively related to the levels of inflammatory TNF-α, IL-1β, and IL-6 as revealed by Spearman's correlation analysis. Moreover, transcription and immunohistochemistry analysis revealed that B. producta D4 and B. producta DSM2950 intervention remarkably suppressed the transcription and expression levels of hepatic Tlr4, MyD88, and caspase-3 (p < 0.05). These data indicated that B. producta may be a good candidate for probiotics in the prevention of ALI.
Collapse
Affiliation(s)
- Bingyong Mao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, People's Republic of China
| | - Weiling Guo
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, People's Republic of China
| | - Xuemei Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, People's Republic of China
| | - Shumao Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, People's Republic of China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, People's Republic of China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, People's Republic of China
| | - Xin Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, People's Republic of China.
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, People's Republic of China.
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, People's Republic of China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, People's Republic of China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122, People's Republic of China
| |
Collapse
|
22
|
Chen F, Li B, Li W, Chen W, Huang Y, Tian Y, Yang B, Yuan M, Xu D, Cao N. Polysaccharide of Atractylodes macrocephala Koidz alleviate lipopolysaccharide-stimulated liver inflammation injury of goslings through miR-223/NLRP3 axis. Poult Sci 2023; 102:102285. [PMID: 36436369 PMCID: PMC9706645 DOI: 10.1016/j.psj.2022.102285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/08/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
Lipopolysaccharide (LPS) infection could cause severe liver inflammation and lead to liver damage, even death. Previous studies have shown that polysaccharide of Atractylodes macrocephala Koidz (PAMK) could protect liver from inflammation caused by LPS in mice. However, whether PAMK could alleviate liver inflammatory injury in other animals with LPS is still unknown. For evaluating whether PAMK could alleviate liver inflammatory injury in goslings with LPS, a total of 80 healthy 1-day old Magang goslings were randomly divided into 4 groups (control group, PAMK group, LPS group, and PAMK+LPS group). Goslings in control group and LPS group were fed with basal diet, and goslings in PAMK group and PAMK+LPS group were fed basal diet supplemented with 400 mg/kg PAMK to the end of trial. On 24 d of age, goslings in the control group and PAMK group were intraperitoneal injected 0.5 mL normal saline, and goslings in LPS and PAMK+LPS groups were intraperitoneal injected with LPS at 5 mg/kg BW. The serum and liver samples were collected for further analysis after treatment of LPS at 6, 12, 24, and 48 h. Furthermore, the hepatocytes were extracted from goose embryo to measure the expression of the key genes of miR-223/NLRP3 axis. The results showed that PAMK pretreatment could maintain normal cell morphology of liver, alleviate the enhanced levels of biochemical indexes ALT and AST, decrease the levels of IL-1β and IL-18, increase the relative mRNA expression of miR-223, and decrease the expression of NLRP3, Caspase-1, and cleaved Caspase-1 in liver and hepatocytes of goslings induced by LPS. These results indicated that PAMK could relieve inflammatory liver tissue damage after LPS treatment and downregulate the level of inflammation factors via miR-223/NLRP3 axis, thus playing a liver protective role in liver inflammation injury in goslings.
Collapse
Affiliation(s)
- Feiyue Chen
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Bingxin Li
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Wanyan Li
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Wenbin Chen
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Yunmao Huang
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Yunbo Tian
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Baohe Yang
- Yunnan Kuaidaduo Animal Husbandry Technology Co., Ltd, Yuxi 653100, China
| | - Mingfeng Yuan
- Yunnan Kuaidaduo Animal Husbandry Technology Co., Ltd, Yuxi 653100, China
| | - Danning Xu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China
| | - Nan Cao
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China; Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Guangzhou 510225, China.
| |
Collapse
|
23
|
Sex-Specific Alterations in Inflammatory MicroRNAs in Mouse Brain and Bone Marrow CD11b+ Cells Following Traumatic Brain Injury. Cell Mol Neurobiol 2023; 43:423-429. [PMID: 34761332 DOI: 10.1007/s10571-021-01164-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/01/2021] [Indexed: 01/12/2023]
Abstract
Sex is a key biological variable in traumatic brain injury (TBI) and plays a significant role in neuroinflammatory responses. However, the molecular mechanisms contributing to this sexually dimorphic neuroinflammatory response remain elusive. Here we describe a significant and previously unreported tissue enrichment and sex-specific alteration of a set of inflammatory microRNAs (miRNAs) in CD11b+ cells of brain and bone marrow isolated from naïve mice as well as mice subjected to TBI. Our data from naïve mice demonstrated that expression levels of miR-146a-5p and miR-150-5p were relatively higher in brain CD11b+ cells, and that miR-155-5p and miR-223-3p were highly enriched in bone marrow CD11b+ cells. Furthermore, while miR-150-5p and miR-155-5p levels were higher in male brain CD11b+ cells, no significant sexual difference was observed for miR-146a-5p and miR-223-3p. However, TBI resulted in sex-specific differential responses of these miRNAs in brain CD11b+ cells. Specifically, miR-223-3p levels in brain CD11b+ cells were markedly elevated in both sexes in response to TBI at 3 and 24 h, with levels in females being significantly higher than males at 24 h. We then focused on analyzing several miR-223-3p targets and inflammation-related marker genes following injury. Corresponding to the greater elevation of miR-223-3p in females, the miR-223-3p targets, TRAF6 and FBXW7 were significantly reduced in females compared to males. Interestingly, anti-inflammatory genes ARG1 and IL4 were higher in females after TBI than in males. These observations suggest miR-223-3p and other inflammatory responsive miRNAs may play a key role in sex-specific neuroinflammatory response following TBI.
Collapse
|
24
|
López‐Cuevas P, Xu C, Severn CE, Oates TCL, Cross SJ, Toye AM, Mann S, Martin P. Macrophage Reprogramming with Anti-miR223-Loaded Artificial Protocells Enhances In Vivo Cancer Therapeutic Potential. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202717. [PMID: 36314048 PMCID: PMC9762313 DOI: 10.1002/advs.202202717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/15/2022] [Indexed: 06/16/2023]
Abstract
Several immune cell-expressed miRNAs (miRs) are associated with altered prognostic outcome in cancer patients, suggesting that they may be potential targets for development of cancer therapies. Here, translucent zebrafish (Danio rerio) is utilized to demonstrate that genetic knockout or knockdown of one such miR, microRNA-223 (miR223), globally or specifically in leukocytes, does indeed lead to reduced cancer progression. As a first step toward potential translation to a clinical therapy, a novel strategy is described for reprogramming neutrophils and macrophages utilizing miniature artificial protocells (PCs) to deliver anti-miRs against the anti-inflammatory miR223. Using genetic and live imaging approaches, it is shown that phagocytic uptake of anti-miR223-loaded PCs by leukocytes in zebrafish (and by human macrophages in vitro) effectively prolongs their pro-inflammatory state by blocking the suppression of pro-inflammatory cytokines, which, in turn, drives altered immune cell-cancer cell interactions and ultimately leads to a reduced cancer burden by driving reduced proliferation and increased cell death of tumor cells. This PC cargo delivery strategy for reprogramming leukocytes toward beneficial phenotypes has implications also for treating other systemic or local immune-mediated pathologies.
Collapse
Affiliation(s)
- Paco López‐Cuevas
- School of BiochemistryBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
| | - Can Xu
- Centre for Protolife ResearchSchool of ChemistryUniversity of BristolBristolBS8 1TSUK
| | - Charlotte E. Severn
- School of BiochemistryBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolBS34 7QHUK
| | - Tiah C. L. Oates
- School of BiochemistryBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolBS34 7QHUK
| | - Stephen J. Cross
- Wolfson Bioimaging FacilityBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
| | - Ashley M. Toye
- School of BiochemistryBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolBS34 7QHUK
| | - Stephen Mann
- Centre for Protolife ResearchSchool of ChemistryUniversity of BristolBristolBS8 1TSUK
- Max Planck Bristol Centre for Minimal BiologySchool of ChemistryUniversity of BristolBristolBS8 1TSUK
- School of Materials Science and EngineeringShanghai Jiao Tong UniversityShanghai200240P. R. China
| | - Paul Martin
- School of BiochemistryBiomedical Sciences BuildingUniversity WalkUniversity of BristolBristolBS8 1TDUK
| |
Collapse
|
25
|
Diaz-Beneitez E, Cubas-Gaona LL, Candelas-Rivera O, Benito-Zafra A, Sánchez-Aparicio MT, Miorin L, Rodríguez JF, García-Sastre A, Rodríguez D. Interaction between chicken TRIM25 and MDA5 and their role in mediated antiviral activity against IBDV infection. Front Microbiol 2022; 13:1068328. [PMID: 36519174 PMCID: PMC9742432 DOI: 10.3389/fmicb.2022.1068328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2023] Open
Abstract
Infectious Bursal Disease Virus (IBDV) is the causative agent of an immunosuppressive disease that affects domestic chickens (Gallus gallus) severely affecting poultry industry worldwide. IBDV infection is characterized by a rapid depletion of the bursal B cell population by apoptosis and the atrophy of this chief lymphoid organ. Previous results from our laboratory have shown that exposure of infected cells to type I IFN leads to an exacerbated apoptosis, indicating an important role of IFN in IBDV pathogenesis. It has been described that recognition of the dsRNA IBDV genome by MDA5, the only known cytoplasmic pattern recognition receptor for viral RNA in chickens, leads to type I IFN production. Here, we confirm that TRIM25, an E3 ubiquitin ligase that leads to RIG-I activation in mammalian cells, significantly contributes to positively regulate MDA5-mediated activation of the IFN-inducing pathway in chicken DF-1 cells. Ectopic expression of chTRIM25 together with chMDA5 or a deletion mutant version exclusively harboring the CARD domains (chMDA5 2CARD) enhances IFN-β and NF-ĸB promoter activation. Using co-immunoprecipitation assays, we show that chMDA5 interacts with chTRIM25 through the CARD domains. Moreover, chTRIM25 co-localizes with both chMDA5 and chMDA5 2CARD, but not with chMDA5 mutant proteins partially or totally lacking these domains. On the other hand, ablation of endogenous chTRIM25 expression reduces chMDA5-induced IFN-β and NF-ĸB promoter activation. Interestingly, ectopic expression of either wild-type chTRIM25, or a mutant version (chTRIM25 C59S/C62S) lacking the E3 ubiquitin ligase activity, restores the co-stimulatory effect of chMDA5 in chTRIM25 knockout cells, suggesting that the E3-ubiquitin ligase activity of chTRIM25 is not required for its downstream IFN-β and NF-ĸB activating function. Also, IBDV-induced expression of IFN-β, Mx and OAS genes was reduced in chTRIM25 knockout as compared to wild-type cells, hence contributing to the enhancement of IBDV replication. Enhanced permissiveness to replication of other viruses, such as avian reovirus, Newcastle disease virus and vesicular stomatitis virus was also observed in chTRIM25 knockout cells. Additionally, chTRIM25 knockout also results in reduced MAVS-induced IFN-β promoter stimulation. Nonetheless, similarly to its mammalian counterpart, chTRIM25 overexpression in wild-type DF-1 cells causes the degradation of ectopically expressed chMAVS.
Collapse
Affiliation(s)
- Elisabet Diaz-Beneitez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | | | - Oscar Candelas-Rivera
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Ana Benito-Zafra
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Maria Teresa Sánchez-Aparicio
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lisa Miorin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - José F. Rodríguez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine, New York, NY, United States
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pathology, Molecular and Cell-Based MedicineI at Mount Sinai, Icahn School of Medicine, New York, NY, United States
| | - Dolores Rodríguez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, CSIC, Madrid, Spain
| |
Collapse
|
26
|
Sharma S, Tyagi T, Antoniak S. Platelet in thrombo-inflammation: Unraveling new therapeutic targets. Front Immunol 2022; 13:1039843. [PMID: 36451834 PMCID: PMC9702553 DOI: 10.3389/fimmu.2022.1039843] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
In the broad range of human diseases, thrombo-inflammation appears as a clinical manifestation. Clinically, it is well characterized in context of superficial thrombophlebitis that is recognized as thrombosis and inflammation of superficial veins. However, it is more hazardous when developed in the microvasculature of injured/inflamed/infected tissues and organs. Several diseases like sepsis and ischemia-reperfusion can cause formation of microvascular thrombosis subsequently leading to thrombo-inflammation. Thrombo-inflammation can also occur in cases of antiphospholipid syndrome, preeclampsia, sickle cell disease, bacterial and viral infection. One of the major contributors to thrombo-inflammation is the loss of normal anti-thrombotic and anti-inflammatory potential of the endothelial cells of vasculature. This manifest itself in the form of dysregulation of the coagulation pathway and complement system, pathologic platelet activation, and increased recruitment of leukocyte within the microvasculature. The role of platelets in hemostasis and formation of thrombi under pathologic and non-pathologic conditions is well established. Platelets are anucleate cells known for their essential role in primary hemostasis and the coagulation pathway. In recent years, studies provide strong evidence for the critical involvement of platelets in inflammatory processes like acute ischemic stroke, and viral infections like Coronavirus disease 2019 (COVID-19). This has encouraged the researchers to investigate the contribution of platelets in the pathology of various thrombo-inflammatory diseases. The inhibition of platelet surface receptors or their intracellular signaling which mediate initial platelet activation and adhesion might prove to be suitable targets in thrombo-inflammatory disorders. Thus, the present review summarizes the concept and mechanism of platelet signaling and briefly discuss their role in sterile and non-sterile thrombo-inflammation, with the emphasis on role of platelets in COVID-19 induced thrombo-inflammation. The aim of this review is to summarize the recent developments in deciphering the role of the platelets in thrombo-inflammation and discuss their potential as pharmaceutical targets.
Collapse
Affiliation(s)
- Swati Sharma
- UNC Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Tarun Tyagi
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Silvio Antoniak
- UNC Blood Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
27
|
Recent advances of nanotechnology in the treatment and diagnosis of polycystic ovary syndrome. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
28
|
Mügge FL, Morlock GE. Planar bioluminescent cytotoxicity assay via genetically modified adherent human reporter cell lines, applied to authenticity screening of Saussurea costus root. J Chromatogr A 2022; 1683:463522. [DOI: 10.1016/j.chroma.2022.463522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 01/02/2023]
|
29
|
Murray A, Banota T, Guo GL, Smith LC, Meshanni JA, Lee J, Kong B, Abramova EV, Goedken M, Gow AJ, Laskin JD, Laskin DL. Farnesoid X receptor regulates lung macrophage activation and injury following nitrogen mustard exposure. Toxicol Appl Pharmacol 2022; 454:116208. [PMID: 35998709 PMCID: PMC9960619 DOI: 10.1016/j.taap.2022.116208] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 02/04/2023]
Abstract
Nitrogen mustard (NM) is a cytotoxic vesicant known to cause acute lung injury which progresses to fibrosis; this is associated with a sequential accumulation of pro- and anti-inflammatory macrophages in the lung which have been implicated in NM toxicity. Farnesoid X receptor (FXR) is a nuclear receptor involved in regulating lipid homeostasis and inflammation. In these studies, we analyzed the role of FXR in inflammatory macrophage activation, lung injury and oxidative stress following NM exposure. Wild-type (WT) and FXR-/- mice were treated intratracheally with PBS (control) or NM (0.08 mg/kg). Bronchoalveolar lavage fluid (BAL) and lung tissue were collected 3, 14 and 28 d later. NM caused progressive histopathologic alterations in the lung including inflammatory cell infiltration and alveolar wall thickening and increases in protein and cells in BAL; oxidative stress was also noted, as reflected by upregulation of heme oxygenase-1. These changes were more prominent in male FXR-/- mice. Flow cytometric analysis revealed that loss of FXR resulted in increases in proinflammatory macrophages at 3 d post NM; this correlated with upregulation of COX-2 and ARL11, markers of macrophage activation. Markers of anti-inflammatory macrophage activation, CD163 and STAT6, were also upregulated after NM; this response was exacerbated in FXR-/- mice at 14 d post-NM. These findings demonstrate that FXR plays a role in limiting macrophage inflammatory responses important in lung injury and oxidative stress. Maintaining or enhancing FXR function may represent a useful strategy in the development of countermeasures to treat mustard lung toxicity.
Collapse
Affiliation(s)
- Alexa Murray
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Tanvi Banota
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Grace L Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Ley Cody Smith
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Jaclynn A Meshanni
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Jordan Lee
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Bo Kong
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Elena V Abramova
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Michael Goedken
- Research Pathology Services, Rutgers University, Piscataway, NJ 08854, USA
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, NJ 08854, USA
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
30
|
Kotlyarov S. High-Density Lipoproteins: A Role in Inflammation in COPD. Int J Mol Sci 2022; 23:8128. [PMID: 35897703 PMCID: PMC9331387 DOI: 10.3390/ijms23158128] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 02/04/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a widespread disease associated with high rates of disability and mortality. COPD is characterized by chronic inflammation in the bronchi as well as systemic inflammation, which contributes significantly to the clinically heterogeneous course of the disease. Lipid metabolism disorders are common in COPD, being a part of its pathogenesis. High-density lipoproteins (HDLs) are not only involved in lipid metabolism, but are also part of the organism's immune and antioxidant defense. In addition, HDL is a versatile transport system for endogenous regulatory agents and is also involved in the removal of exogenous substances such as lipopolysaccharide. These functions, as well as information about lipoprotein metabolism disorders in COPD, allow a broader assessment of their role in the pathogenesis of heterogeneous and comorbid course of the disease.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
31
|
Farmanzadeh A, Qujeq D, Yousefi T. The Interaction Network of MicroRNAs with Cytokines and Signaling Pathways in Allergic Asthma. Microrna 2022; 11:104-117. [PMID: 35507792 DOI: 10.2174/2211536611666220428134324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/15/2022] [Accepted: 03/10/2022] [Indexed: 01/01/2023]
Abstract
Allergic asthma is a complicated disease that is affected by many factors. Numerous cytokines and signaling pathways are attributed to the cause of asthma symptoms. MicroRNAs (miRNAs) are a group of small non-coding single-stranded RNA molecules that are involved in gene silencing and posttranscriptional regulation of gene expression by targeting mRNAs. In pathological conditions, altered expression of microRNAs differentially regulates cytokines and signaling pathways and therefore, can be the underlying reason for the pathogenesis of allergic asthma. Indeed, microRNAs participate in airway inflammation via inducing airway structural cells and activating immune responses by targeting cytokines and signaling pathways. Thus, to make a complete understanding of allergic asthma, it is necessary to investigate the communication network of microRNAs with cytokines and signaling pathways which is contributed to the pathogenesis of allergic asthma. Here, we shed light on this aspect of asthma pathology by Summarizing our current knowledge of this topic.
Collapse
Affiliation(s)
- Ali Farmanzadeh
- Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Tooba Yousefi
- Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
32
|
Chang Y, Syahirah R, Oprescu SN, Wang X, Jung J, Cooper SH, Torregrosa-Allen S, Elzey BD, Hsu AY, Randolph LN, Sun Y, Kuang S, Broxmeyer HE, Deng Q, Lian X, Bao X. Chemically-defined generation of human hemogenic endothelium and definitive hematopoietic progenitor cells. Biomaterials 2022; 285:121569. [PMID: 35567999 PMCID: PMC10065832 DOI: 10.1016/j.biomaterials.2022.121569] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/23/2022] [Accepted: 05/03/2022] [Indexed: 12/17/2022]
Abstract
Human hematopoietic stem cells (HSCs), which arise from aorta-gonad-mesonephros (AGM), are widely used to treat blood diseases and cancers. However, a technique for their robust generation in vitro is still missing. Here we show temporal manipulation of Wnt signaling is sufficient and essential to induce AGM-like hematopoiesis from human pluripotent stem cells. TGFβ inhibition at the stage of aorta-like SOX17+CD235a- hemogenic endothelium yielded AGM-like hematopoietic progenitors, which closely resembled primary cord blood HSCs at the transcriptional level and contained diverse lineage-primed progenitor populations via single cell RNA-sequencing analysis. Notably, the resulting definitive cells presented lymphoid and myeloid potential in vitro; and could home to a definitive hematopoietic site in zebrafish and rescue bloodless zebrafish after transplantation. Engraftment and multilineage repopulating activities were also observed in mouse recipients. Together, our work provided a chemically-defined and feeder-free culture platform for scalable generation of AGM-like hematopoietic progenitor cells, leading to enhanced production of functional blood and immune cells for various therapeutic applications.
Collapse
Affiliation(s)
- Yun Chang
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN, 47907, USA
| | - Ramizah Syahirah
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Stephanie N Oprescu
- Purdue University Center for Cancer Research, West Lafayette, IN, 47907, USA; Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Xuepeng Wang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Juhyung Jung
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN, 47907, USA
| | - Scott H Cooper
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | | | - Bennett D Elzey
- Purdue University Center for Cancer Research, West Lafayette, IN, 47907, USA; Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, 47907, USA
| | - Alan Y Hsu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Department of Pathology, Harvard Medical School, Boston, MA, 02115, USA
| | - Lauren N Randolph
- Departments of Biomedical Engineering, Biology, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yufei Sun
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Shihuan Kuang
- Purdue University Center for Cancer Research, West Lafayette, IN, 47907, USA; Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Qing Deng
- Purdue University Center for Cancer Research, West Lafayette, IN, 47907, USA; Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| | - Xiaojun Lian
- Departments of Biomedical Engineering, Biology, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.
| | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN, 47907, USA.
| |
Collapse
|
33
|
Jiang Y, Xu X, Xiao L, Wang L, Qiang S. The Role of microRNA in the Inflammatory Response of Wound Healing. Front Immunol 2022; 13:852419. [PMID: 35386721 PMCID: PMC8977525 DOI: 10.3389/fimmu.2022.852419] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/02/2022] [Indexed: 12/20/2022] Open
Abstract
Wound healing, a highly complex pathophysiological response to injury, includes four overlapping phases of hemostasis, inflammation, proliferation, and remodeling. Initiation and resolution of the inflammatory response are the primary requirements for wound healing, and are also key events that determines wound quality and healing time. Currently, the number of patients with persistent chronic wounds has generally increased, which imposes health and economic burden on patients and society. Recent studies have found that microRNA(miRNA) plays an essential role in the inflammation involved in wound healing and may provide a new therapeutic direction for wound treatment. Therefore, this review focused on the role and significance of miRNA in the inflammation phase of wound healing.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Xiang Xu
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Long Xiao
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Lihong Wang
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Sheng Qiang
- Center Laboratory, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| |
Collapse
|
34
|
Bernareggi D, Xie Q, Prager BC, Yun J, Cruz LS, Pham TV, Kim W, Lee X, Coffey M, Zalfa C, Azmoon P, Zhu H, Tamayo P, Rich JN, Kaufman DS. CHMP2A regulates tumor sensitivity to natural killer cell-mediated cytotoxicity. Nat Commun 2022; 13:1899. [PMID: 35393416 PMCID: PMC8990014 DOI: 10.1038/s41467-022-29469-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 03/09/2022] [Indexed: 02/08/2023] Open
Abstract
Natural killer (NK) cells are known to mediate killing of various cancer types, but tumor cells can develop resistance mechanisms to escape NK cell-mediated killing. Here, we use a "two cell type" whole genome CRISPR-Cas9 screening system to discover key regulators of tumor sensitivity and resistance to NK cell-mediated cytotoxicity in human glioblastoma stem cells (GSC). We identify CHMP2A as a regulator of GSC resistance to NK cell-mediated cytotoxicity and we confirm these findings in a head and neck squamous cells carcinoma (HNSCC) model. We show that deletion of CHMP2A activates NF-κB in tumor cells to mediate increased chemokine secretion that promotes NK cell migration towards tumor cells. In the HNSCC model we demonstrate that CHMP2A mediates tumor resistance to NK cells via secretion of extracellular vesicles (EVs) that express MICA/B and TRAIL. These secreted ligands induce apoptosis of NK cells to inhibit their antitumor activity. To confirm these in vitro studies, we demonstrate that deletion of CHMP2A in CAL27 HNSCC cells leads to increased NK cell-mediated killing in a xenograft immunodeficient mouse model. These findings illustrate a mechanism of tumor immune escape through EVs secretion and identify inhibition of CHMP2A and related targets as opportunities to improve NK cell-mediated immunotherapy.
Collapse
Affiliation(s)
- Davide Bernareggi
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Qi Xie
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.,Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China
| | - Briana C Prager
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.,Cleveland Clinic Lerner College of Medicine at Cleveland Clinic & Case Western Reserve University, Cleveland, OH, USA
| | - Jiyoung Yun
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Luisjesus S Cruz
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Timothy V Pham
- Center for Novel Therapeutics and Moores Cancer Center, UCSD, San Diego, CA, USA
| | - William Kim
- Center for Novel Therapeutics and Moores Cancer Center, UCSD, San Diego, CA, USA.,Division of Medical Genetics, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Xiqing Lee
- Department of Oncology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Michael Coffey
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cristina Zalfa
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Pardis Azmoon
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Huang Zhu
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Pablo Tamayo
- Center for Novel Therapeutics and Moores Cancer Center, UCSD, San Diego, CA, USA.,Division of Medical Genetics, Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Jeremy N Rich
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Dan S Kaufman
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
35
|
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV) and the closely related SARS-CoV-2 are emergent highly pathogenic human respiratory viruses causing acute lethal disease associated with lung damage and dysregulated inflammatory responses. SARS-CoV envelope protein (E) is a virulence factor involved in the activation of various inflammatory pathways. Here, we study the contribution of host miRNAs to the virulence mediated by E protein. Small RNAseq analysis of infected mouse lungs identified miRNA-223 as a potential regulator of pulmonary inflammation, since it was significantly increased in SARS-CoV-WT virulent infection compared to the attenuated SARS-CoV-ΔE infection. In vivo inhibition of miRNA-223-3p increased mRNA levels of pro-inflammatory cytokines and NLRP3 inflammasome, suggesting that during lung infection, miRNA-223 might contribute to restrict an excessive inflammatory response. Interestingly, miRNA-223-3p inhibition also increased the levels of the CFTR transporter, which is involved in edema resolution and was significantly downregulated in the lungs of mice infected with the virulent SARS-CoV-WT virus. At the histopathological level, a decrease in the pulmonary edema was observed when miR-223-3p was inhibited, suggesting that miRNA-223-3p was involved in the regulation of the SARS-CoV-induced inflammatory pathology. These results indicate that miRNA-223 participates in the regulation of E protein-mediated inflammatory response during SARS-CoV infection by targeting different host mRNAs involved in the pulmonary inflammation, and identify miRNA-223 as a potential therapeutic target in SARS-CoV infection.
Collapse
|
36
|
Brasil Brandao B, Lino M, Kahn CR. Extracellular miRNAs as mediators of obesity-associated disease. J Physiol 2022; 600:1155-1169. [PMID: 34392542 PMCID: PMC8845532 DOI: 10.1113/jp280910] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular miRNAs are found in a variety of body fluids and mediate intercellular and interorgan communication, thus regulating gene expression and cellular metabolism. These miRNAs are secreted either in small vesicles/exosomes (sEV) or bound to proteins such as Argonaute and high-density lipoprotein. Both exosomal and protein-bound circulating miRNAs are altered in obesity. Although all tissues can contribute to changes in circulating miRNAs, adipose tissue itself is an important source of these miRNAs, especially those in sEVs. These are derived from both adipocytes and macrophages and participate in crosstalk between these cells, as well as peripheral tissues, including liver, skeletal muscle and pancreas, whose function may be impaired in obesity. Changes in levels of circulating miRNAs have also been linked to the beneficial effects induced by weight loss interventions, including diet, exercise and bariatric surgery, further indicating a role for these miRNAs as mediators of disease pathogenesis. Here, we review the role of circulating miRNAs in the pathophysiology of obesity and explore their potential use as biomarkers and in therapy of obesity-associated metabolic syndrome.
Collapse
Affiliation(s)
- Bruna Brasil Brandao
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | - Marsel Lino
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| | - C. Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
37
|
Yao F, Deng Y, Zhao Y, Mei Y, Zhang Y, Liu X, Martinez C, Su X, Rosato RR, Teng H, Hang Q, Yap S, Chen D, Wang Y, Chen MJM, Zhang M, Liang H, Xie D, Chen X, Zhu H, Chang JC, You MJ, Sun Y, Gan B, Ma L. A targetable LIFR-NF-κB-LCN2 axis controls liver tumorigenesis and vulnerability to ferroptosis. Nat Commun 2021; 12:7333. [PMID: 34921145 PMCID: PMC8683481 DOI: 10.1038/s41467-021-27452-9] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/16/2021] [Indexed: 02/06/2023] Open
Abstract
The growing knowledge of ferroptosis has suggested the role and therapeutic potential of ferroptosis in cancer, but has not been translated into effective therapy. Liver cancer, primarily hepatocellular carcinoma (HCC), is highly lethal with limited treatment options. LIFR is frequently downregulated in HCC. Here, by studying hepatocyte-specific and inducible Lifr-knockout mice, we show that loss of Lifr promotes liver tumorigenesis and confers resistance to drug-induced ferroptosis. Mechanistically, loss of LIFR activates NF-κB signaling through SHP1, leading to upregulation of the iron-sequestering cytokine LCN2, which depletes iron and renders insensitivity to ferroptosis inducers. Notably, an LCN2-neutralizing antibody enhances the ferroptosis-inducing and anticancer effects of sorafenib on HCC patient-derived xenograft tumors with low LIFR expression and high LCN2 expression. Thus, anti-LCN2 therapy is a promising way to improve liver cancer treatment by targeting ferroptosis.
Collapse
Affiliation(s)
- Fan Yao
- Hubei Hongshan Laboratory, College of Life Science and Technology, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Yalan Deng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yang Zhao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ying Mei
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yilei Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaoguang Liu
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Consuelo Martinez
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xiaohua Su
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Roberto R Rosato
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Hongqi Teng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Qinglei Hang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shannon Yap
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dahu Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yumeng Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mei-Ju May Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mutian Zhang
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX, 77030, USA
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dong Xie
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Hao Zhu
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jenny C Chang
- Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - M James You
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yutong Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA.
| |
Collapse
|
38
|
Madrigal T, Hernández-Monge J, Herrera LA, González-De la Rosa CH, Domínguez-Gómez G, Candelaria M, Luna-Maldonado F, Calderón González KG, Díaz-Chávez J. Regulation of miRNAs Expression by Mutant p53 Gain of Function in Cancer. Front Cell Dev Biol 2021; 9:695723. [PMID: 34957087 PMCID: PMC8697023 DOI: 10.3389/fcell.2021.695723] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 11/15/2021] [Indexed: 12/19/2022] Open
Abstract
The p53 roles have been largely described; among them, cell proliferation and apoptosis control are some of the best studied and understood. Interestingly, the mutations on the six hotspot sites within the region that encodes the DNA-binding domain of p53 give rise to other very different variants. The particular behavior of these variants led to consider p53 mutants as separate oncogene entities; that is, they do not retain wild type functions but acquire new ones, namely Gain-of-function p53 mutants. Furthermore, recent studies have revealed how p53 mutants regulate gene expression and exert oncogenic effects by unbalancing specific microRNAs (miRNAs) levels that provoke epithelial-mesenchymal transition, chemoresistance, and cell survival, among others. In this review, we discuss recent evidence of the crosstalk between miRNAs and mutants of p53, as well as the consequent cellular processes dysregulated.
Collapse
Affiliation(s)
- Tzitzijanik Madrigal
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Mexico City, Mexico
- Departamento de Ciencias Biológicas y de La Salud, UAM Iztapalapa, Mexico City, Mexico
| | - Jesús Hernández-Monge
- Cátedra-CONACyT Laboratorio de Biomarcadores Moleculares, Instituto de Física, UASLP, San Luis Potosí, Mexico
| | - Luis A Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Mexico City, Mexico
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | | | | | - Myrna Candelaria
- Subdirección de Investigación Clínica, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Fernando Luna-Maldonado
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Karla G Calderón González
- Laboratorio de Interacciones Biomoleculares y Cáncer, Instituto de Física, UASLP, San Luis Potosi, Mexico
| | - José Díaz-Chávez
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Mexico City, Mexico
| |
Collapse
|
39
|
Circulating miRNAs as Potential Biomarkers Distinguishing Relapsing-Remitting from Secondary Progressive Multiple Sclerosis. A Review. Int J Mol Sci 2021; 22:ijms222111887. [PMID: 34769314 PMCID: PMC8584709 DOI: 10.3390/ijms222111887] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/30/2021] [Accepted: 10/31/2021] [Indexed: 12/18/2022] Open
Abstract
Multiple sclerosis (MS) is a debilitating neurodegenerative, highly heterogeneous disease with a variable course. The most common MS subtype is relapsing–remitting (RR), having interchanging periods of worsening and relative stabilization. After a decade, in most RR patients, it alters into the secondary progressive (SP) phase, the most debilitating one with no clear remissions, leading to progressive disability deterioration. Among the greatest challenges for clinicians is understanding disease progression molecular mechanisms, since RR is mainly characterized by inflammatory processes, while in SP, the neurodegeneration prevails. This is especially important because distinguishing RR from the SP subtype early will enable faster implementation of appropriate treatment. Currently, the MS course is not well-correlated with the biomarkers routinely used in clinical practice. Despite many studies, there are still no reliable indicators correlating with the disease stage and its activity degree. Circulating microRNAs (miRNAs) may be considered valuable molecules for the MS diagnosis and, presumably, helpful in predicting disease subtype. MiRNA expression dysregulation is commonly observed in the MS course. Moreover, knowledge of diverse miRNA panel expression between RRMS and SPMS may allow for deterring disability progression through successful treatment. Therefore, in this review, we address the current state of research on differences in miRNA panel expression between the phases.
Collapse
|
40
|
Roffel MP, Maes T, Brandsma CA, van den Berge M, Vanaudenaerde BM, Joos GF, Brusselle GG, Heijink IH, Bracke KR. MiR-223 is increased in lungs of patients with COPD and modulates cigarette smoke-induced pulmonary inflammation. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1091-L1104. [PMID: 34668437 DOI: 10.1152/ajplung.00252.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Since microRNA (miR)-223-3p modulates inflammatory responses and COPD is associated with amplified pulmonary inflammation, we hypothesized that miR-223-3p plays a role in COPD pathogenesis. Expression of miR-223-3p was measured in lung tissue of 2 independent cohorts with COPD GOLD stage II-IV patients, never smokers and smokers without COPD. The functional role of miR-223-3p was studied in deficient mice and upon overexpression in airway epithelial cells from COPD and controls. We observed higher miR-223-3p levels in patients with COPD stage II-IV compared to (non)-smoking controls, and levels were associated with higher neutrophil numbers in bronchial biopsies of COPD patients. MiR-223-3p expression was also increased in lungs and bronchoalveolar lavage of cigarette smoke (CS)-exposed mice. CS-induced neutrophil and monocyte lung infiltration was stronger in miR-223 deficient mice upon acute (5 days) exposure, but attenuated upon sub-chronic (4 weeks) exposure. Additionally, miR-223 deficiency attenuated acute and sub-chronic CS-induced lung infiltration of dendritic cells and T lymphocytes. Finally, in vitro overexpression of miR-223-3p in non-COPD airway epithelial cells suppressed CXCL8 and GM-CSF secretion and gene expression of the pro-inflammatory transcription factor TRAF6. Importantly, this suppressive effect of miR-223-3p was compromised in COPD-derived cultures. In conclusion, we demonstrate that miR-223-3p is increased in lungs of COPD patients and CS-exposed mice, and is associated with neutrophilic inflammation. In vivo data indicate that miR-223 acts as negative regulator of acute CS-induced neutrophilic and monocytic inflammation. In vitro data suggests that miR-223-3p does so by suppressing pro-inflammatory airway epithelial responses, which is less effective in COPD epithelium.
Collapse
Affiliation(s)
- Mirjam P Roffel
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, The Netherlands.,Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| | - Tania Maes
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| | - Corry-Anke Brandsma
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, The Netherlands
| | - Maarten van den Berge
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, The Netherlands.,University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands
| | - Bart M Vanaudenaerde
- Laboratory for Respiratory Diseases, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Guy F Joos
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| | - Guy G Brusselle
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, The Netherlands.,University of Groningen, University Medical Center Groningen, Department of Pulmonology, Groningen, The Netherlands
| | - Ken R Bracke
- Ghent University, Ghent University Hospital, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent, Belgium
| |
Collapse
|
41
|
Liu W, Hsu AY, Wang Y, Lin T, Sun H, Pachter JS, Groisman A, Imperioli M, Yungher FW, Hu L, Wang P, Deng Q, Fan Z. Mitofusin-2 regulates leukocyte adhesion and β2 integrin activation. J Leukoc Biol 2021; 111:771-791. [PMID: 34494308 DOI: 10.1002/jlb.1a0720-471r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Neutrophils are critical for inflammation and innate immunity, and their adhesion to vascular endothelium is a crucial step in neutrophil recruitment. Mitofusin-2 (MFN2) is required for neutrophil adhesion, but molecular details are unclear. Here, we demonstrated that β2 -integrin-mediated slow-rolling and arrest, but not PSGL-1-mediated cell rolling, are defective in MFN2-deficient neutrophil-like HL60 cells. This adhesion defect is associated with reduced expression of fMLP (N-formylmethionyl-leucyl-phenylalanine) receptor FPR1 as well as the inhibited β2 integrin activation, as assessed by conformation-specific monoclonal antibodies. MFN2 deficiency also leads to decreased actin polymerization, which is important for β2 integrin activation. Mn2+ -induced cell spreading is also inhibited after MFN2 knockdown. MFN2 deficiency limited the maturation of β2 integrin activation during the neutrophil-directed differentiation of HL60 cells, which is indicated by CD35 and CD87 markers. MFN2 knockdown in β2-integrin activation-matured cells (CD87high population) also inhibits integrin activation, indicating that MFN2 directly affects β2 integrin activation. Our study illustrates the function of MFN2 in leukocyte adhesion and may provide new insights into the development and treatment of MFN2 deficiency-related diseases.
Collapse
Affiliation(s)
- Wei Liu
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - Alan Y Hsu
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Yueyang Wang
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Tao Lin
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - Hao Sun
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Joel S Pachter
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - Alex Groisman
- Department of Physics, University of California San Diego, La Jolla, California, USA
| | | | | | - Liang Hu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Penghua Wang
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - Qing Deng
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA.,Purdue Institute for Inflammation, Immunology, & Infectious Disease, Purdue University, West Lafayette, Indiana, USA.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut, USA
| |
Collapse
|
42
|
Abuelezz NZ, E Shabana M, Rashed L, Nb Morcos G. Nanocurcumin Modulates miR-223-3p and NF-κB Levels in the Pancreas of Rat Model of Polycystic Ovary Syndrome to Attenuate Autophagy Flare, Insulin Resistance and Improve ß Cell Mass. J Exp Pharmacol 2021; 13:873-888. [PMID: 34475786 PMCID: PMC8405883 DOI: 10.2147/jep.s323962] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose Polycystic ovary syndrome (PCOS) is a prevalent female endocrine disorder. 50–70% of PCOS patients suffer from glucose intolerance, insulin and β cell impairments. Updated studies reveal the crucial regulatory role of inflammation modulators in various diseases, by manipulating autophagy and oxidative stress. However, the data available about autophagy in PCOS pancreas, especially in relation to inflammation key players are little. This study investigated pancreatic autophagy status in PCOS rat model, with miR-223-3p and NF-κB levels as pivotal regulators of oxidative stress-autophagy axis, insulin, and β cell integrity. We then analyzed nanocurcumin effects as a putative anti-inflammatory nutraceutical on the disrupted parameters. Methods Nanocurcumin was characterized using transmission electron microscopy (TEM) and Fourier-transform IR (FT-IR) spectroscopy. Adult virgin Wistar rats were selected, and PCOS was induced using letrozole (1mg/kg). Nanocurcumin was ingested following letrozole. Sex hormones and insulin resistance were determined. miR-223-3p expression was determined using real-time PCR. Immunohistochemistry and Western blotting determined β cells, NF-κB, and autophagy markers p62 and LC3II. Results PCOS group showed significant disruptions in sex hormones and a double fold increase in glucose and insulin levels, exhibiting insulin resistance. Immunostaining confirmed around 46% deterioration of ß cell mass. Real-time PCR showed significant downregulation of miR-223-3p. Immunohistochemistry and Western blotting revealed a drastic upsurge of NF-κB, and autophagy markers p62 and LC3II, confirming bioinformatics target analysis. Interestingly, compared to PCOS group, nanocurcumin (200mg/kg) significantly upregulated miR-223-3p expression by 30%. It subsided NF-κB and autophagy eruption to restore ß cell mass and attenuate insulin resistance. Conclusion To the best of our knowledge, this study is the first to highlight the vital contribution of miR-223-3p and NF-κB levels in aggravating PCOS pancreatic autophagy and consequent impairments. It spots nanocurcumin potential as an inflammation and autophagy modulator, for possible better management of PCOS complications.
Collapse
Affiliation(s)
- Nermeen Z Abuelezz
- Biochemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, Giza, Egypt
| | - Marwa E Shabana
- Pathology Department, Medical Division, National Research Center, Cairo, Egypt
| | - Laila Rashed
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - George Nb Morcos
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt.,Basic Medical Science Department, Faculty of Medicine, King Salman International University, El Tur, Egypt
| |
Collapse
|
43
|
Jiao P, Wang XP, Luoreng ZM, Yang J, Jia L, Ma Y, Wei DW. miR-223: An Effective Regulator of Immune Cell Differentiation and Inflammation. Int J Biol Sci 2021; 17:2308-2322. [PMID: 34239357 PMCID: PMC8241730 DOI: 10.7150/ijbs.59876] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/21/2021] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs) play a critical role in regulating various biological processes, such as cell differentiation and immune modulation by binding to their target genes. miR-223 is a miRNA with important functions and has been widely investigated in recent years. Under certain physiological conditions, miR-223 is regulated by different transcription factors, including sirtuin1 (Sirt1), PU.1 and Mef2c, and its biological functions are mediated through changes in its cellular or tissue expression. This review paper summarizes miR-223 biosynthesis and its regulatory role in the differentiation of granulocytes, dendritic cells (DCs) and lymphocytes, macrophage polarization, and endothelial and epithelial inflammation. In addition, it describes the molecular mechanisms of miR-223 in regulating lung inflammation, rheumatoid arthritis, enteritis, neuroinflammation and mastitis to provide insights into the existing molecular regulatory networks and therapies for inflammatory diseases in humans and animals.
Collapse
Affiliation(s)
- Peng Jiao
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Xing-Ping Wang
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Zhuo-Ma Luoreng
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Jian Yang
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Li Jia
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Yun Ma
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| | - Da-Wei Wei
- School of Agriculture, Ningxia University, Yinchuan 750021, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan 750021, China
| |
Collapse
|
44
|
Wu YZ, Chan KYY, Leung KT, Lam HS, Tam YH, Lee KH, Li K, Ng PC. The miR-223/nuclear factor I-A axis regulates inflammation and cellular functions in intestinal tissues with necrotizing enterocolitis. FEBS Open Bio 2021; 11:1907-1920. [PMID: 33932136 PMCID: PMC8255851 DOI: 10.1002/2211-5463.13164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/05/2021] [Accepted: 04/12/2021] [Indexed: 01/01/2023] Open
Abstract
We previously demonstrated that microRNA(miR)‐223 is overexpressed in intestinal tissue of infants with necrotizing enterocolitis (NEC). The objective of the current study was to identify the target gene of miR‐223 and to investigate the role of the miR‐223/nuclear factor I‐A (NFIA) axis in cellular functions that underpin the pathophysiology of NEC. The target gene of miR‐223 was identified by in silico target prediction bioinformatics, luciferase assay, and western blotting. We investigated downstream signals of miR‐223 and cellular functions by overexpressing the miRNA in Caco‐2 and FHs74 cells stimulated with lipopolysaccharide or lipoteichoic acid (LTA). NFIA was identified as a target gene of miR‐223. Overexpression of miR‐223 significantly induced MYOM1 and inhibited NFIA and RGN in Caco‐2 cells, while costimulation with LTA decreased expression of GNA11, MYLK, and PRKCZ. Expression levels of GNA11, MYLK, IL‐6, and IL‐8 were increased, and levels of NFIA and RGN were decreased in FHs74 cells. These potential downstream genes were significantly correlated with levels of miR‐223 or NFIA in primary NEC tissues. Overexpression of miR‐223 significantly increased apoptosis of Caco‐2 and FHs74 cells, while proliferation of FHs74 was inhibited. These results suggest that upon binding with NFIA, miR‐223 regulates functional effectors in pathways of apoptosis, cell proliferation, G protein signaling, inflammation, and smooth muscle contraction. The miR‐223/NFIA axis may play an important role in the pathophysiology of NEC by enhancing inflammation and tissue damage.
Collapse
Affiliation(s)
- Yu Zheng Wu
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Kathy Yuen Yee Chan
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Kam Tong Leung
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Hugh Simon Lam
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Yuk Him Tam
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kim Hung Lee
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Karen Li
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Pak Cheung Ng
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
45
|
Sullivan C, Soos BL, Millard PJ, Kim CH, King BL. Modeling Virus-Induced Inflammation in Zebrafish: A Balance Between Infection Control and Excessive Inflammation. Front Immunol 2021; 12:636623. [PMID: 34025644 PMCID: PMC8138431 DOI: 10.3389/fimmu.2021.636623] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
The inflammatory response to viral infection in humans is a dynamic process with complex cell interactions that are governed by the immune system and influenced by both host and viral factors. Due to this complexity, the relative contributions of the virus and host factors are best studied in vivo using animal models. In this review, we describe how the zebrafish (Danio rerio) has been used as a powerful model to study host-virus interactions and inflammation by combining robust forward and reverse genetic tools with in vivo imaging of transparent embryos and larvae. The innate immune system has an essential role in the initial inflammatory response to viral infection. Focused studies of the innate immune response to viral infection are possible using the zebrafish model as there is a 4-6 week timeframe during development where they have a functional innate immune system dominated by neutrophils and macrophages. During this timeframe, zebrafish lack a functional adaptive immune system, so it is possible to study the innate immune response in isolation. Sequencing of the zebrafish genome has revealed significant genetic conservation with the human genome, and multiple studies have revealed both functional conservation of genes, including those critical to host cell infection and host cell inflammatory response. In addition to studying several fish viruses, zebrafish infection models have been developed for several human viruses, including influenza A, noroviruses, chikungunya, Zika, dengue, herpes simplex virus type 1, Sindbis, and hepatitis C virus. The development of these diverse viral infection models, coupled with the inherent strengths of the zebrafish model, particularly as it relates to our understanding of macrophage and neutrophil biology, offers opportunities for far more intensive studies aimed at understanding conserved host responses to viral infection. In this context, we review aspects relating to the evolution of innate immunity, including the evolution of viral pattern recognition receptors, interferons and interferon receptors, and non-coding RNAs.
Collapse
Affiliation(s)
- Con Sullivan
- College of Arts and Sciences, University of Maine at Augusta, Bangor, ME, United States
| | - Brandy-Lee Soos
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, United States
| | - Paul J. Millard
- Department of Environmental and Sustainable Engineering, University at Albany, Albany, NY, United States
| | - Carol H. Kim
- Department of Biomedical Sciences, University at Albany, Albany, NY, United States
- Department of Biological Sciences, University at Albany, Albany, NY, United States
| | - Benjamin L. King
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME, United States
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| |
Collapse
|
46
|
Chen L, Zhu X, Han B, Ji L, Yao L, Wang Z. High Expression of microRNA-223 Indicates a Good Prognosis in Triple-Negative Breast Cancer. Front Oncol 2021; 11:630432. [PMID: 33928027 PMCID: PMC8078593 DOI: 10.3389/fonc.2021.630432] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/12/2021] [Indexed: 12/19/2022] Open
Abstract
Purpose MicroRNAs can influence many biological processes and have shown promise as cancer biomarkers. Few studies have focused on the expression of microRNA-223 (miR-223) and its precise role in breast cancer (BC). We aimed to examine the expression level of miR-223 and its prognostic value in BC. Methods Tissue microarray (TMA)-based miRNA detection in situ hybridization (ISH) with a locked nucleic acid (LNA) probe was used to detect miR-223 expression in 450 BC tissue samples. Overall survival (OS) and disease-free survival (DFS) were compared between two groups using the Kaplan-Meier method and Cox regression model. Results OS and DFS were prolonged in the high miR-223 expression group compared to the low miR-223 expression group (p < 0.0001 and p = 0.017, respectively), especially in patients with the triple-negative breast cancer (TNBC) subtype (p = 0.046 and p < 0.001, respectively). Univariate and multivariate Cox regression analyses revealed that TNM stage (p = 0.008), the molecular subtype (p = 0.049), and miR-223 (p < 0.001) were independently associated with OS and DFS. External validation was performed with the METABRIC and The Cancer Genome Atlas (TCGA) databases via online webtools and was consistent with the data described above. Conclusions This study provides evidence that high miR-223 expression at diagnosis is associated with improved DFS and OS for BC patients, especially those with the TNBC subtype. miR-223 is a valid and independent prognostic biomarker in BC.
Collapse
Affiliation(s)
- Li Chen
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiuzhi Zhu
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Boyue Han
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Ji
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling Yao
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhonghua Wang
- Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
47
|
Wright K, de Silva K, Plain KM, Purdie AC, Blair TA, Duggin IG, Britton WJ, Oehlers SH. Mycobacterial infection-induced miR-206 inhibits protective neutrophil recruitment via the CXCL12/CXCR4 signalling axis. PLoS Pathog 2021; 17:e1009186. [PMID: 33826679 PMCID: PMC8055004 DOI: 10.1371/journal.ppat.1009186] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/19/2021] [Accepted: 03/29/2021] [Indexed: 12/22/2022] Open
Abstract
Pathogenic mycobacteria actively dysregulate protective host immune signalling pathways during infection to drive the formation of permissive granuloma microenvironments. Dynamic regulation of host microRNA (miRNA) expression is a conserved feature of mycobacterial infections across host-pathogen pairings. Here we examine the role of miR-206 in the zebrafish model of Mycobacterium marinum infection, which allows investigation of the early stages of granuloma formation. We find miR-206 is upregulated following infection by pathogenic M. marinum and that antagomir-mediated knockdown of miR-206 is protective against infection. We observed striking upregulation of cxcl12a and cxcr4b in infected miR-206 knockdown zebrafish embryos and live imaging revealed enhanced recruitment of neutrophils to sites of infection. We used CRISPR/Cas9-mediated knockdown of cxcl12a and cxcr4b expression and AMD3100 inhibition of Cxcr4 to show that the enhanced neutrophil response and reduced bacterial burden caused by miR-206 knockdown was dependent on the Cxcl12/Cxcr4 signalling axis. Together, our data illustrate a pathway through which pathogenic mycobacteria induce host miR-206 expression to suppress Cxcl12/Cxcr4 signalling and prevent protective neutrophil recruitment to granulomas.
Collapse
Affiliation(s)
- Kathryn Wright
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
- The University of Sydney, Faculty of Science, Sydney School of Veterinary Science, Sydney, New South Wales, Australia
| | - Kumudika de Silva
- The University of Sydney, Faculty of Science, Sydney School of Veterinary Science, Sydney, New South Wales, Australia
| | - Karren M. Plain
- The University of Sydney, Faculty of Science, Sydney School of Veterinary Science, Sydney, New South Wales, Australia
| | - Auriol C. Purdie
- The University of Sydney, Faculty of Science, Sydney School of Veterinary Science, Sydney, New South Wales, Australia
| | - Tamika A. Blair
- ithree Institute, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Iain G. Duggin
- ithree Institute, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Warwick J. Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Stefan H. Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
- The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, New South Wales, Australia
| |
Collapse
|
48
|
Zhong S, Golpon H, Zardo P, Borlak J. miRNAs in lung cancer. A systematic review identifies predictive and prognostic miRNA candidates for precision medicine in lung cancer. Transl Res 2021; 230:164-196. [PMID: 33253979 DOI: 10.1016/j.trsl.2020.11.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 11/05/2020] [Accepted: 11/24/2020] [Indexed: 02/08/2023]
Abstract
Lung cancer (LC) is the leading cause of cancer-related death worldwide and miRNAs play a key role in LC development. To better diagnose LC and to predict drug treatment responses we evaluated 228 articles encompassing 16,697 patients and 12,582 healthy controls. Based on the criteria of ≥3 independent studies and a sensitivity and specificity of >0.8 we found blood-borne miR-20a, miR-10b, miR-150, and miR-223 to be excellent diagnostic biomarkers for non-small cell LC whereas miR-205 is specific for squamous cell carcinoma. The systematic review also revealed 38 commonly regulated miRNAs in tumor tissue and the circulation, thus enabling the prediction of histological subtypes of LC. Moreover, theranostic biomarker candidates with proven responsiveness to checkpoint inhibitor treatments were identified, notably miR-34a, miR-93, miR-106b, miR-181a, miR-193a-3p, and miR-375. Conversely, miR-103a-3p, miR-152, miR-152-3p, miR-15b, miR-16, miR-194, miR-34b, and miR-506 influence programmed cell death-ligand 1 and programmed cell death-1 receptor expression, therefore providing a rationale for the development of molecularly targeted therapies. Furthermore, miR-21, miR-25, miR-27b, miR-19b, miR-125b, miR-146a, and miR-210 predicted response to platinum-based treatments. We also highlight controversial reports on specific miRNAs. In conclusion, we report diagnostic miRNA biomarkers for in-depth clinical evaluation. Furthermore, in an effort to avoid unnecessary toxicity we propose predictive biomarkers. The biomarker candidates support personalized treatment decisions of LC patients and await their confirmation in randomized clinical trials.
Collapse
Affiliation(s)
- Shen Zhong
- Centre for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| | - Heiko Golpon
- Department of Pneumology, Hannover Medical School, Hannover, Germany
| | - Patrick Zardo
- Clinic for Cardiothoracic and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
49
|
Wang X, Chi J, Dong B, Xu L, Zhou Y, Huang Y, Sun S, Wei F, Liu Y, Liu C, Che K, Lv W, Chen Y, Wang Y. MiR-223-3p and miR-22-3p inhibit monosodium urate-induced gouty inflammation by targeting NLRP3. Int J Rheum Dis 2021; 24:599-607. [PMID: 33650318 DOI: 10.1111/1756-185x.14089] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/20/2021] [Accepted: 02/06/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND MicroRNAs (miRNAs) have been shown to play a crucial role in inflammation regulation; however, their relationship with inflammation in acute gouty arthritis has not been fully elucidated. Herein, we conducted a study to explore the regulatory roles of miR-223-3p and miR-22-3p in gouty-associated inflammation. METHODS In vitro and in vivo experiments were conducted to examine the molecular mechanisms of miRNA regulation in gouty inflammation. Dual-luciferase reporter assay was used to verify the direct target of miR-223-3p and miR-22-3p. RESULTS We found that miR-223-3p and miR-22-3p interacted with the 3' untranslated region segment of NLRP3 (nucleotide-binding domain leucine-rich repeat [NLR] and pyrin domain containing receptor 3) and inhibited its expression. A decreased expression of miR-223-3p and miR-22-3p was observed in both mice air pouch synovium and phorbol myristrate acetate-treated THP-1 cells stimulated with monosodium urate (P < .05). Compared with the negative control group, NLRP3 expression at the transcript and protein level in miR-223-3p and miR-22-3p overexpression group significantly decreased after 6 hours of monosodium urate treatment in vivo and in vitro (P < .05). The results of the dual-luciferase reporter assay demonstrated that miR-223-3p and miR-22-3p directly targeted NLRP3. CONCLUSION The findings of the present study show that miR-223-3p and miR-22-3p can reduce the inflammatory effects of gout by inhibiting the expression of NLRP3.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
- Transfusion Section, Chongqing University Three Gorges Hospital, Qingdao, China
| | - Jingwei Chi
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Bingzi Dong
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Lili Xu
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Yue Zhou
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Yajing Huang
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Shengnan Sun
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Fanxiang Wei
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Yuzhao Liu
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Chuanfeng Liu
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Kui Che
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Wenshan Lv
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Ying Chen
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Yangang Wang
- Department of Endocrinology, Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| |
Collapse
|
50
|
Chen B, Han J, Chen S, Xie R, Yang J, Zhou T, Zhang Q, Xia R. MicroLet-7b Regulates Neutrophil Function and Dampens Neutrophilic Inflammation by Suppressing the Canonical TLR4/NF-κB Pathway. Front Immunol 2021; 12:653344. [PMID: 33868293 PMCID: PMC8044834 DOI: 10.3389/fimmu.2021.653344] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/04/2021] [Indexed: 12/25/2022] Open
Abstract
Sepsis is a heterogeneous syndrome caused by a dysregulated host response during the process of infection. Neutrophils are involved in the development of sepsis due to their essential role in host defense. COVID-19 is a viral sepsis. Disfunction of neutrophils in sepsis has been described in previous studies, however, little is known about the role of microRNA-let-7b (miR-let-7b), toll-like receptor 4 (TLR4), and nuclear factor kappa B (NF-κB) activity in neutrophils and how they participate in the development of sepsis. In this study, we investigated the regulatory pathway of miR-let-7b/TLR4/NF-κB in neutrophils. We also explored the downstream cytokines released by neutrophils following miR-let-7b treatment and its therapeutic effects in cecal ligation and puncture (CLP)-induced septic mice. Six-to-eight-week-old male C57BL/6 mice underwent CLP following treatment with miR-let-7b agomir. Survival (n=10), changes in liver and lungs histopathology (n=4), circulating neutrophil counts (n=4), the liver-body weight ratio (n=4–7), and the lung wet-to-dry ratio (n=5–6) were recorded. We found that overexpression of miR-let-7b could significantly down-regulate the expression of human-derived neutrophilic TLR4 at a post-transcriptional level, a decreased level of proinflammatory factors including interleukin-6 (IL-6), IL-8, tumor necrosis factor α (TNF-α), and an upregulation of anti-inflammatory factor IL-10 in vitro. After miR-let-7b agomir treatment in vivo, neutrophil recruitment was inhibited and thus the injuries of liver and lungs in CLP-induced septic mice were alleviated (p=0.01 and p=0.04, respectively), less weight loss was reduced, and survival in septic mice was also significantly improved (p=0.013). Our study suggested that miR-let-7b could be a potential target of sepsis.
Collapse
Affiliation(s)
- Binzhen Chen
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Jia Han
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Shaoheng Chen
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Rufeng Xie
- Blood Engineering Laboratory, Shanghai Blood Center, Shanghai, China
| | - Jie Yang
- Blood Engineering Laboratory, Shanghai Blood Center, Shanghai, China
| | - Tongming Zhou
- Shanghai Key Laboratory of Data Science, School of Computer Science, Fudan University, Shanghai, China
| | - Qi Zhang
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| | - Rong Xia
- Department of Blood Transfusion, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|