1
|
Zamudio-Beltrán LE, Bossu CM, Bueno-Hernández AA, Dunn PO, Sly ND, Rayne C, Anderson EC, Hernández-Baños BE, Ruegg KC. Parallel and convergent evolution in genes underlying seasonal migration. Evol Lett 2025; 9:189-208. [PMID: 40191407 PMCID: PMC11968193 DOI: 10.1093/evlett/qrae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 04/09/2025] Open
Abstract
Seasonal migration has fascinated scientists and natural historians for centuries. While the genetic basis of migration has been widely studied across different taxa, there is little consensus regarding which genomic regions play a role in the ability to migrate and whether they are similar across species. Here, we examine the genetic basis of intraspecific variation within and between distinct migratory phenotypes in a songbird. We focus on the Common Yellowthroat (Geothlypis trichas) as a model system because the polyphyletic origin of eastern and western clades across North America provides a strong framework for understanding the extent to which there has been parallel or convergent evolution in the genes associated with migratory behavior. First, we investigate genome-wide population genetic structure in the Common Yellowthroat in 196 individuals collected from 22 locations across breeding range. Then, to identify candidate genes involved in seasonal migration, we identify signals of putative selection in replicate comparisons between resident and migratory phenotypes within and between eastern and western clades. Overall, we find wide-spread support for parallel evolution at the genic level, particularly in genes that mediate biological timekeeping. However, we find little evidence of parallelism at the individual SNP level, supporting the idea that there are multiple genetic pathways involved in the modulation of migration.
Collapse
Affiliation(s)
- Luz E Zamudio-Beltrán
- Facultad de Estudios Superiores Zaragoza, UNAM, Mexico City, Mexico
- Facultad de Ciencias, UNAM, Mexico City, Mexico
| | - Christen M Bossu
- Department of Biology, Colorado State University, Fort Collins, CO, United States
| | | | - Peter O Dunn
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Nicholas D Sly
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, United States
| | - Christine Rayne
- Department of Biology, Colorado State University, Fort Collins, CO, United States
| | - Eric C Anderson
- Department of Biology, Colorado State University, Fort Collins, CO, United States
| | | | - Kristen C Ruegg
- Department of Biology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
2
|
Liang KH, Chen YC, Hsu CY, Kao ZK, Tsai PH, Huang HY, Chu YC, Ho HL, Liao YC, Lee YC, Huang CC, Wei TC, Liao YJ, Lu YH, Kuo CT, Chiou SH. Predictive biosignatures for hospitalization in patients with virologically confirmed COVID-19. J Chin Med Assoc 2025; 88:246-252. [PMID: 39730204 DOI: 10.1097/jcma.0000000000001203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, presents with varying severity among individuals. Both viral and host factors can influence the severity of acute and chronic COVID-19, with chronic COVID-19 commonly referred to as long COVID. SARS-CoV-2 infection can be properly diagnosed by performing real-time reverse transcription polymerase chain reaction analysis of nasal swab samples. Pulse oximetry, chest X-ray, and complete blood count (CBC) analysis can be used to assess the condition of the patient to ensure that the appropriate medical care is delivered. This study aimed to develop biosignatures that can be used to distinguish between patients who are likely to develop severe disease and require hospitalization from patients who can be safely monitored in less intensive settings. METHODS A retrospective investigation was conducted on 7897 adult patients with virologically confirmed SARS-CoV-2 infection between January 26, 2020, and November 30, 2023; all patients underwent comprehensive CBC testing at Taipei Veterans General Hospital. Among them, 1867 patients were independently recruited for a population study involving genome-wide genotyping of approximately 424 000 genomic variants. Therefore, the participants were divided into two patient cohorts, one with genomic data (n = 1867) and one without (n = 6030) for model validation and training, respectively. RESULTS We constructed and validated a biosignature model by using a combination of CBC measurements to predict subsequent hospitalization events (hazard ratio = 3.38, 95% confidence interval: 3.07-3.73 for the training cohort and 3.03 [2.46-3.73] for the validation cohort; both p < 10 -8 ). The obtained scores were used to identify the top quartile of patients, who formed the "very high risk" group with a significantly higher cumulative incidence of hospitalization (log-rank p < 10 -8 in both the training and validation cohorts). The "very high risk" group exhibited a cumulative hospitalization rate of >60%, whereas the rate for the other patients was approximately 30% over a 1.5-year period, providing a binary classification of patients with distinct hospitalization risks. To investigate the genetic factors mediating this risk, we conducted a genome-wide association study. Specific regions in chromosomes 7 and 10 and the mitochondrial chromosome (M), harboring IKAROS family zinc finger 1 ( IKZF1 ), actin binding LIM protein 1 ( ABLIM1 ), and mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 3 ( MT-ND3 ), exhibited prominent associations with binary risk classification. The identified exonic variants of IKZF1 are linked to several autoimmune diseases. Notably, people with different genotypes of the leading variants (rs4132601, rs141492519, and Affx-120744614) exhibited varying cumulative hospitalization rates after infection. CONCLUSION We successfully developed and validated a biosignature model of COVID-19 severe disease in virologically confirmed patients. The identified genomic variants provide new insights for infectious disease research and medical care.
Collapse
Affiliation(s)
- Kung-Hao Liang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Biosafety Level 3 Laboratory, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- College of Medicine, Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- School of Pharmaceutical Sciences, Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yu-Chun Chen
- Department of Family Medicine, Taipei Veterans General Hospital, Yuli Branch, Hualien, Taiwan, ROC
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chun-Yi Hsu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Zih-Kai Kao
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Hsin-Yi Huang
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yuan-Chia Chu
- Department of Medical Research, Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Information Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan, ROC
| | - Hsiang-Ling Ho
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yi-Chu Liao
- Departments of Neurology, Taipei Veterans General Hospital, Taiwan, ROC
- Department of Neurology, School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan, ROC
- College of Medicine, Brain Research Center, National Yang Ming Chao Tung University School of Medicine, Taipei, Taiwan, ROC
| | - Yi-Chung Lee
- Departments of Neurology, Taipei Veterans General Hospital, Taiwan, ROC
- Department of Neurology, School of Medicine, National Yang Ming Chao Tung University, Taipei, Taiwan, ROC
- College of Medicine, Brain Research Center, National Yang Ming Chao Tung University School of Medicine, Taipei, Taiwan, ROC
| | - Chi-Cheng Huang
- Department of Surgery, Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Division of Breast Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- College of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Tzu-Chun Wei
- Department of Urology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Urology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- College of Medicine, Shu-Tien Urological Institute, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yi-Jia Liao
- Departments of Neurology, Taipei Veterans General Hospital, Taiwan, ROC
| | - Yung-Hsiu Lu
- Department of Pediatrics, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chen-Tsung Kuo
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
3
|
Wei Q, Niu ZC, Fu XY, OuYang MH, Guo XT, Li J, Chang YX, Mao HP, Gao XM. S6K/FLNC/ITGβ3 signaling pathway regulates osteoclastogenesis and the inhibition of osteoclastogenesis by columbianadin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156276. [PMID: 39612888 DOI: 10.1016/j.phymed.2024.156276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 10/31/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Over-activation of osteoclastogenesis is a significant factor contributing to bone loss, leading to increased resorption of bone. Columbianadin (CBN), a compound derived from Angelicae Pubescentis Radix, has traditionally been used in Chinese medicine to treat bone-related disorders. However, the specific effects of CBN on bone loss are still poorly understood. STUDY DESIGN This study aims to identify a novel target for inhibiting osteoclast differentiation and to elucidate the effects and underlying mechanisms of CBN on osteoclastogenesis. METHODS We employed a transcriptomics approach to identify genes that undergo significant changes during osteoclast differentiation. These findings were validated using reverse transcription-polymerase chain reaction (RT-PCR) and Western blot (WB) analysis. Subsequently, we utilized adenoviral transfection to investigate the effects of target genes on osteoclast differentiation. Additionally, we employed proteomics to elucidate the signaling pathways that regulate osteoclast differentiation. We examined the effect of CBN on S6K/FLNC/ITGβ3 signaling pathway and osteoclast differentiation. RESULTS Our results revealed a dramatic increase in filamin C (FLNC) levels during osteoclast differentiation. Inhibition of FLNC expression significantly suppressed markers of osteoclast differentiation such as tartrate-resistant acid phosphatase (TRAP), nuclear factor-activated T cell 1 (NFATc1), and c-Fos, as well as inhibited the activity of bone resorption. We further conducted a proteomic analysis and found S6 K protein might be involved in this process. Then we utilized an S6K-specific inhibitor (PF-4,708,671) and demonstrated that inhibiting the S6 K protein reduced FLNC expression and the interaction between FLNC and integrin β3 (ITGβ3) in osteoclasts. Finally, we found that CBN inhibited osteoclast differentiation and bone loss in ovariectomized mice by targeting the S6K/FLNC/ITGβ3 signaling pathway. CONCLUSION FLNC was identified as a critical protein in osteoclastogenesis. The S6K/FLNC/ITGβ3 signaling pathway played a significant role in osteoclast differentiation and bone loss. Furthermore, CBN exhibited anti-osteoporotic effects by inhibiting the S6K/FLNC/ITGβ3 signaling pathway.
Collapse
Affiliation(s)
- Qiu Wei
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, PR China
| | - Zi-Chang Niu
- The First Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, PR China
| | - Xiao-Yu Fu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, PR China
| | - Ming-Hui OuYang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, PR China
| | - Xiao-Tong Guo
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, PR China
| | - Jin Li
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, PR China
| | - Yan-Xu Chang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| | - Hao-Ping Mao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, PR China.
| | - Xiu-Mei Gao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, PR China.
| |
Collapse
|
4
|
Tang T, Wu M, Yang L, Liu F, Zhang F. Muscle LIM protein of Macrobrachium nipponense (MnMLP) involved in immune and stress response. FISH & SHELLFISH IMMUNOLOGY 2024; 153:109809. [PMID: 39122098 DOI: 10.1016/j.fsi.2024.109809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
The muscle LIM protein (MLP) is a member of the cysteine and glycine-rich protein (CSRP) family, composed of CSRP1, CSRP2 and CSRP3/MLP. MLP is involved in a multitude of functional roles, including cytoskeletal organization, transcriptional regulation, and signal transduction. However, the molecular mechanisms underlying its involvement in immune and stress responses remain to be elucidated. This study identified an MnMLP in the freshwater crustacean Macrobrachium nipponense. The isothermal titration calorimetry assay demonstrated that recombinant MnMLP was capable of coordinating with Zn2+. Upon challenge by Aeromonas veronii or WSSV, and exposure to CdCl2, up-regulation was recorded in the muscle and intestinal tissues, suggesting its involvement in immune and anti-stress responses. MnMLP protein was predominantly expressed in the cytoplasm of the transfected HEK-293T cells, but after treatment with LPS, Cd2+ or H2O2, the MnMLP was observed to be transferred into the nucleus. The comet assay demonstrated that the overexpression of MnMLP could mitigate the DNA damage induced by H2O2 in HEK-293T cells, suggesting the potential involvement of MnMLP in the DNA repair process. These findings suggest that DNA repair may represent a possible mechanism by which MnMLP may be involved in the host's defense against pathogens and stress.
Collapse
Affiliation(s)
- Ting Tang
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Mengjia Wu
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Likun Yang
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Fengsong Liu
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China; Hebei Basic Science Center for Biotic Interaction, Hebei University, Baoding, 071002, China; Engineering Research Center of Ecological Safety and Conservation in Beijing-Tianjin-Hebei (Xiong'an New Area) of MOE, Baoding, 071002, China.
| | - Feng Zhang
- Key Laboratory of Zoological Systematics and Application, College of Life Sciences, Hebei University, Baoding, 071002, China; Hebei Basic Science Center for Biotic Interaction, Hebei University, Baoding, 071002, China.
| |
Collapse
|
5
|
Qiu Y, Yang M, Che X, Yu X, Zhi K. miR-2467-3p/ABLIM1 Axis Mediates the Formation and Progression of Deep Vein Thrombosis by Regulating Inflammation and Oxidative Stress. Int J Angiol 2024; 33:174-181. [PMID: 39131807 PMCID: PMC11315604 DOI: 10.1055/s-0044-1779663] [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: 08/13/2024] Open
Abstract
Deep vein thrombosis (DVT) is a common postoperative complication of orthopaedic surgery with a complex pathogenesis mechanism. The effect of the miR-2467-3p/acting-binding LIM protein 1 (ABLIM1) axis on thrombus formation and human vascular endothelial cells (HUVECs) progression was evaluated aiming to identify a novel potential biomarker of DVT. DVT rat models were established by inferior vena cava stenosis. The expression of the miR-2467-3p/ABLIM1 axis was analyzed by PCR. HUVECs were induced with oxidative low-density lipoprotein (ox-LDL). Cell growth and motility were assessed by cell counting kit 8 (CCK8) and Transwell assay. The inflammation and oxidative stress were estimated by proinflammatory cytokines and generation of MDA and reactive oxygen species (ROS). ABLIM1 was downregulated in DVT rats. Overexpressing ABLIM1 could suppress the formation of thrombosis and alleviate inflammation and oxidative stress. In HUVECs, ox-LDL induced significantly increased miR-2467-3p and decreased ABLIM1, and miR-2467-3p could negatively regulate ABLIM1. The knockdown of miR-2467-3p could alleviate the inhibited cell growth and motility by ox-LDL, and the inflammation and oxidative stress were also attenuated. While silencing could reverse the effect of miR-2467-3p on ox-LDL-induced HUVECs. The miR-2467-3p/ABLIM1 axis regulates the occurrence and development of DVT through modulating HUVECs inflammation and oxidative stress.
Collapse
Affiliation(s)
- Yu Qiu
- Department of Vascular and Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Meiying Yang
- Department of Trauma Orthopedics, Zibo Central Hospital, Zibo, China
| | - Xinting Che
- Department of Vascular and Endovascular Surgery, School of Medicine, Xiamen University, Xiamen, China
| | - Xinming Yu
- Department of Vascular Surgery, Zibo Central Hospital, Zibo, China
| | - Kangkang Zhi
- Department of Vascular and Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
6
|
Wang H, Li T, Jiang Y, Chen S, Zou S, Bonewald LF, Duan P. Force-Loaded Cementocytes Regulate Osteoclastogenesis via S1P/S1PR1/Rac1 Axis. J Dent Res 2023; 102:1376-1386. [PMID: 37735908 DOI: 10.1177/00220345231195765] [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] [Indexed: 09/23/2023] Open
Abstract
Orthodontically induced inflammatory root resorption (OIIRR) is the major iatrogenic complication of orthodontic treatment, seriously endangering tooth longevity and impairing masticatory function. Osteoclasts are thought to be the primary effector cells that initiate the pathological process of OIIRR; however, the cellular and molecular mechanisms responsible for OIIRR remain unclear. Our previous studies revealed that cementocytes, the major mechanically responsive cells in cementum, respond to compressive stress to activate and influence osteoclasts locally. For this study, we hypothesized that the sphingosine-1-phosphate (S1P) signaling pathway, a key mechanotransduction pathway in cementocytes, may regulate osteoclasts under the different magnitudes of either physiologic compressive stress that causes tooth movement or pathologic stress that causes OIIRR. Here, we show a biphasic effect of higher compression force stimulating the synthesis and secretion of S1P, whereas lower compression force reduced signaling in IDG-CM6 cementocytes. Using conditioned media from force-loaded cementocytes, we verified the cell-to-cell communication between cementocytes and osteoclasts and show that selective knockdown of S1PR1 and Rac1 plays a role in cementocyte-driven osteoclastogenesis via the S1P/S1PR1/Rac1 axis. Most importantly, the use of inhibitors of this axis reduced or prevented the pathological process of OIIRR. The intercellular communication mechanisms between cementocytes and osteoclasts may serve as a promising therapeutic target for OIIRR.
Collapse
Affiliation(s)
- H Wang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases;Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - T Li
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases;Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology
| | - Y Jiang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases;Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - S Chen
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases;Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - S Zou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases;Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - L F Bonewald
- Departments of Anatomy, Cell Biology & Physiology and Orthopaedic Surgery, Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - P Duan
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases;Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Omata Y, Tachibana H, Aizaki Y, Mimura T, Sato K. Essentiality of Nfatc1 short isoform in osteoclast differentiation and its self-regulation. Sci Rep 2023; 13:18797. [PMID: 37914750 PMCID: PMC10620225 DOI: 10.1038/s41598-023-45909-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023] Open
Abstract
During osteoclast differentiation, the expression of the transcription factor nuclear factor of activated T cell 1 (Nfatc1) increases in an autoproliferative manner. Nfatc1 isoforms are of three sizes, and only the short isoform increases during osteoclast differentiation. Genetic ablation of the whole Nfatc1 gene demonstrated that it is essential for osteoclastogenesis; however, the specific role of the Nfatc1 short form (Nfatc1/αA) remains unknown. In this study, we engineered Nfatc1 short form-specific knockout mice and found that these mice died in utero by day 13.5. We developed a novel osteoclast culture system in which hematopoietic stem cells were cultured, proliferated, and then differentiated into osteoclasts in vitro. Using this system, we show that the Nfatc1/αA isoform is essential for osteoclastogenesis and is responsible for the expression of various osteoclast markers, the Nfatc1 short form itself, and Nfatc1 regulators.
Collapse
Affiliation(s)
- Yasuhiro Omata
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hideyuki Tachibana
- Department of Rheumatology, Akiru Municipal Medical Center, 78-1 Hikita, Akiruno, Tokyo, 197-0834, Japan
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, 38 Moroyama, Iruma, Saitama, 350-0495, Japan
| | - Yoshimi Aizaki
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, 38 Moroyama, Iruma, Saitama, 350-0495, Japan
| | - Toshihide Mimura
- Department of Rheumatology and Applied Immunology, Faculty of Medicine, Saitama Medical University, 38 Moroyama, Iruma, Saitama, 350-0495, Japan
| | - Kojiro Sato
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| |
Collapse
|
8
|
He Z, Liu R, Wang M, Wang Q, Zheng J, Ding J, Wen J, Fahey AG, Zhao G. Combined effect of microbially derived cecal SCFA and host genetics on feed efficiency in broiler chickens. MICROBIOME 2023; 11:198. [PMID: 37653442 PMCID: PMC10472625 DOI: 10.1186/s40168-023-01627-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 07/18/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Improving feed efficiency is the most important goal for modern animal production. The regulatory mechanisms of controlling feed efficiency traits are extremely complex and include the functions related to host genetics and gut microbiota. Short-chain fatty acids (SCFAs), as significant metabolites of microbiota, could be used to refine the combined effect of host genetics and gut microbiota. However, the association of SCFAs with the gut microbiota and host genetics for regulating feed efficiency is far from understood. RESULTS In this study, 464 broilers were housed for RFI measuring and examining the host genome sequence. And 300 broilers were examined for cecal microbial data and SCFA concentration. Genome-wide association studies (GWAS) showed that four out of seven SCFAs had significant associations with genome variants. One locus (chr4: 29414391-29417189), located near or inside the genes MAML3, SETD7, and MGST2, was significantly associated with propionate and had a modest effect on feed efficiency traits and the microbiota. The genetic effect of the top SNP explained 8.43% variance of propionate. Individuals with genotype AA had significantly different propionate concentrations (0.074 vs. 0.131 μg/mg), feed efficiency (FCR: 1.658 vs. 1.685), and relative abundance of 14 taxa compared to those with the GG genotype. Christensenellaceae and Christensenellaceae_R-7_group were associated with feed efficiency, propionate concentration, the top SNP genotypes, and lipid metabolism. Individuals with a higher cecal abundance of these taxa showed better feed efficiency and lower concentrations of caecal SCFAs. CONCLUSION Our study provides strong evidence of the pathway that host genome variants affect the cecal SCFA by influencing caecal microbiota and then regulating feed efficiency. The cecal taxa Christensenellaceae and Christensenellaceae_R-7_group were identified as representative taxa contributing to the combined effect of host genetics and SCFAs on chicken feed efficiency. These findings provided strong evidence of the combined effect of host genetics and gut microbial SCFAs in regulating feed efficiency traits. Video Abstract.
Collapse
Affiliation(s)
- Zhengxiao He
- State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Ranran Liu
- State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Mengjie Wang
- State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Qiao Wang
- State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jumei Zheng
- State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jiqiang Ding
- State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jie Wen
- State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Alan G Fahey
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland.
| | - Guiping Zhao
- State Key Laboratory of Animal Nutrition; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
9
|
Prasansuklab A, Sukjamnong S, Theerasri A, Hu VW, Sarachana T, Tencomnao T. Transcriptomic analysis of glutamate-induced HT22 neurotoxicity as a model for screening anti-Alzheimer's drugs. Sci Rep 2023; 13:7225. [PMID: 37142620 PMCID: PMC10160028 DOI: 10.1038/s41598-023-34183-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023] Open
Abstract
Glutamate-induced neurotoxicity in the HT22 mouse hippocampal neuronal cell line has been recognized as a valuable cell model for the study of neurotoxicity associated with neurodegenerative diseases including Alzheimer's disease (AD). However, the relevance of this cell model for AD pathogenesis and preclinical drug screening remains to be more elucidated. While there is increasing use of this cell model in a number of studies, relatively little is known about its underlying molecular signatures in relation to AD. Here, our RNA sequencing study provides the first transcriptomic and network analyses of HT22 cells following glutamate exposure. Several differentially expressed genes (DEGs) and their relationships specific to AD were identified. Additionally, the usefulness of this cell model as a drug screening system was assessed by determining the expression of those AD-associated DEGs in response to two medicinal plant extracts, Acanthus ebracteatus and Streblus asper, that have been previously shown to be protective in this cell model. In summary, the present study reports newly identified AD-specific molecular signatures in glutamate-injured HT22 cells, suggesting that this cell can be a valuable model system for the screening and evaluation of new anti-AD agents, particularly from natural products.
Collapse
Affiliation(s)
- Anchalee Prasansuklab
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- College of Public Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Suporn Sukjamnong
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- SYstems Neuroscience of Autism and PSychiatric Disorders (SYNAPS) Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Atsadang Theerasri
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Valerie W Hu
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Tewarit Sarachana
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- SYstems Neuroscience of Autism and PSychiatric Disorders (SYNAPS) Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
10
|
Actin-Binding LIM 1 (ABLIM1) Inhibits Glioblastoma Progression and Serves as a Novel Prognostic Biomarker. DISEASE MARKERS 2022; 2022:9516808. [PMID: 36583064 PMCID: PMC9794427 DOI: 10.1155/2022/9516808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
Background Glioma is the most prevalent malignant brain tumor in adult humans, and glioblastoma (GBM) is the most malignant type. The actin-binding LIM 1 (ABLIM1) protein can modulate actin polymerization, which is essential for the cell proliferation and migration. We aim to investigate ABLIM1 expression, function, and clinical significance in GBM. Methods The ABLIM1 mRNA level was extracted from the TCGA and GTEx online databases. The ABLIM1 protein expression level was explored using immunohistochemistry staining in a GBM cohort enrolled in our hospital (n = 104). The patient survival and prognostic factors were determined using the Kaplan-Meier method and multivariate Cox hazard proportional analysis, respectively. Two human GBM cell lines, U87 and U251 cells, were utilized for ABLIM1 overexpression and cell proliferation analyses. A subcutaneous xenograft model was generated using nude mice to validate the tumor-related effect of ABLIM1 in vivo. Results ABLIM1 exhibited a significantly lower mRNA level in GBM than in other glioma or normal brain tissues. Higher ABLIM1 protein level was correlated with smaller GBM tumor size and better cancer-specific survival (CSS). Multivariate analysis identified ABLIM1 as a novel independent prognostic factor for GBM prognosis. ABLIM1 overexpression significantly inhibits U87 and U251 cell proliferation and colony formation. Consistently, ABLIM1 exerted tumor-suppressing functions in mice models. Conclusion ABLIM1 plays antitumor roles in GBM progression and could be served as a novel biomarker to help predict GBM prognosis.
Collapse
|
11
|
Lee HI, Lee GR, Lee J, Kim N, Kwon M, Kim HJ, Kim NY, Park JH, Jeong W. Dehydrocostus lactone inhibits NFATc1 via regulation of IKK, JNK, and Nrf2, thereby attenuating osteoclastogenesis. BMB Rep 2021. [PMID: 31964469 PMCID: PMC7196184 DOI: 10.5483/bmbrep.2020.53.4.220] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Excessive and hyperactive osteoclast activity causes bone diseases such as osteoporosis and periodontitis. Thus, the regulation of osteoclast differentiation has clinical implications. We recently reported that dehydrocostus lactone (DL) inhibits osteoclast differentiation by regulating a nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1), but the underlying mechanism remains to be elucidated. Here we demonstrated that DL inhibits NFATc1 by regulating nuclear factor-κB (NF-κB), activator protein-1 (AP-1), and nuclear factor-erythroid 2-related factor 2 (Nrf2). DL attenuated IκBα phosphorylation and p65 nuclear translocation as well as decreased the expression of NF-κB target genes and c-Fos. It also inhibited c-Jun N-terminal kinase (JNK) but not p38 or extracellular signal-regulated kinase. The reporter assay revealed that DL inhibits NF-κB and AP-1 activation. In addition, DL reduced reactive oxygen species either by scavenging them or by activating Nrf2. The DL inhibition of NFATc1 expression and osteoclast differentiation was less effective in Nrf2-deficient cells. Collectively, these results suggest that DL regulates NFATc1 by inhibiting NF-κB and AP-1 via down-regulation of IκB kinase and JNK as well as by activating Nrf2, and thereby attenuates osteoclast differentiation.
Collapse
Affiliation(s)
- Hye In Lee
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Gong-Rak Lee
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Jiae Lee
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Narae Kim
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Minjeong Kwon
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Hyun Jin Kim
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Nam Young Kim
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Jin Ha Park
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| | - Woojin Jeong
- Department of Life Science and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
12
|
Dong X, Feng M, Yang H, Liu H, Guo H, Gao X, Liu Y, Liu R, Zhang N, Chen R, Kong R. Rictor promotes cell migration and actin polymerization through regulating ABLIM1 phosphorylation in Hepatocellular Carcinoma. Int J Biol Sci 2020; 16:2835-2852. [PMID: 33061800 PMCID: PMC7545703 DOI: 10.7150/ijbs.46285] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/18/2020] [Indexed: 01/01/2023] Open
Abstract
As one of the most ominous malignancies, hepatocellular carcinoma (HCC) is frequently diagnosed at an advanced stage, owing to its aggressive invasion and metastatic spread. Emerging evidence has demonstrated that Rictor, as a unique component of the mTORC2, plays a role in cell migration, as it is dysregulated in various cancers, including HCC. However, the underlying molecular mechanism has not been well-characterized. Here, evaluation on a tissue-array panel and bioinformatics analysis revealed that Rictor is highly expressed in HCC tissues. Moreover, increased Rictor expression predicts poor survival of HCC patients. Rictor knockdown significantly suppressed cell migration and actin polymerization, thereby leading to decreased nuclear accumulation of MKL1 and subsequent inactivation of SRF/MKL1-dependent gene transcription, i.e. Arp3 and c-Fos. Mechanistically, we identified ABLIM1 as a previously unknown phosphorylation target of Rictor. Rictor interacts with ABLIM1 and regulates its serine phosphorylation in HCC cells. We generated ABLIM1 knockout cell lines of HCC, in which dominant negative mutations of Ser 214 and Ser 431 residues inhibited the ABLIM1-mediated actin polymerization and the MKL1 signaling pathway. Overall, ABLIM1 phosphorylation induced by Rictor plays an important role in controlling actin polymerization in HCC cells.
Collapse
Affiliation(s)
- Xin Dong
- Translational Cancer Research Center, Peking University First Hospital, Beijing. 100034, P.R. China
| | - Mei Feng
- Translational Cancer Research Center, Peking University First Hospital, Beijing. 100034, P.R. China.,Department of General Surgery, Peking University First Hospital, Beijing. 100034, P.R. China
| | - Hui Yang
- Translational Cancer Research Center, Peking University First Hospital, Beijing. 100034, P.R. China
| | - Hengkang Liu
- Translational Cancer Research Center, Peking University First Hospital, Beijing. 100034, P.R. China
| | - Hua Guo
- Laboratory of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, P.R. China
| | - Xianshu Gao
- Department of Radiation Oncology, Peking University First Hospital, Beijing. 100034, P.R. China
| | - Yucun Liu
- Department of General Surgery, Peking University First Hospital, Beijing. 100034, P.R. China
| | - Rong Liu
- Translational Cancer Research Center, Peking University First Hospital, Beijing. 100034, P.R. China
| | - Ning Zhang
- Translational Cancer Research Center, Peking University First Hospital, Beijing. 100034, P.R. China
| | - Ruibing Chen
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, P.R. China
| | - Ruirui Kong
- Translational Cancer Research Center, Peking University First Hospital, Beijing. 100034, P.R. China
| |
Collapse
|
13
|
Chen X, Huang Y, Shi D, Nie C, Luo Y, Guo L, Zou Y, Xie C. LncRNA ZNF667-AS1 Promotes ABLIM1 Expression by Adsorbing micro RNA-1290 to Suppress Nasopharyngeal Carcinoma Cell Progression. Onco Targets Ther 2020; 13:4397-4409. [PMID: 32606725 PMCID: PMC7248807 DOI: 10.2147/ott.s245554] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/15/2020] [Indexed: 12/11/2022] Open
Abstract
Background Recently, long non-coding RNAs (lncRNAs) have been elucidated to play essential roles in cancers, and the recognition of lncRNA expression patterns in nasopharyngeal carcinoma (NPC) may be helpful for indicating novel mechanisms underlying NPC carcinogenesis. Herein, we conducted this study to probe into the function of lncRNA ZNF667-AS1 in NPC progression with the involvement of microRNA-1290 (miR-1290) and actin-binding LIM protein 1 (ABLIM1). Materials and Methods In silico analysis screened differentially expressed genes and miRNAs in NPC and predicted potential mechanisms. ZNF667-AS1 expression was detected in NPC tissues and cells. The gain-and-loss function assays were performed to explore the effects of lncRNA ZNF667-AS1 and miR-1290 in NPC cell biological behaviors. In vivo experiments were further conducted to confirm the in vitro results. Results In silico analysis predicted that ZNF667-AS1 was diminished in NPC, which may downregulate ABLIM1 through sponging miR-1290. ZNF667-AS1 was poorly expressed in NPC tissues and cells, and overexpression of ZNF667-AS1 inhibited growth of NPC cells. ZNF667-AS1 competitively bound with miR-1290, thereby upregulating ABLIM1. miR-1290 resulted in the promotion of NPC cell progression by suppressing ABLIM1. Overexpression of ZNF667-AS1 or suppression of miR-1290 inhibited tumorigenicity of NPC cells in vivo. Conclusion This study highlights that lncRNA ZNF667-AS1 promotes ABLIM1 expression by sponging miR-1290 to suppress NPC cell progression.
Collapse
Affiliation(s)
- Xi Chen
- Department of Otorhinolaryngology, Guangdong Women and Children Hospital, Guang Zhou, Guangdong, 511400, People's Republic of China.,Department of Otorhinolaryngology, People's Hospital of Longhua,Guangdong,People's Republic of China
| | - Yaping Huang
- Department of Otorhinolaryngology, Guangdong Women and Children Hospital, Guang Zhou, Guangdong, 511400, People's Republic of China
| | - Dianyu Shi
- Department of Otorhinolaryngology, People's Hospital of Longhua,Guangdong,People's Republic of China
| | - Chuan Nie
- Department of Neonatology, Guangdong Women and Children Hospital, Guang Zhou 511400, Guangdong, People's Republic of China
| | - Yiping Luo
- Department of Internal Medicine, Guangdong Women and Children Hospital, Guang Zhou 511400, Guangdong, People's Republic of China
| | - Liangfen Guo
- Department of Otorhinolaryngology, Guangdong Women and Children Hospital, Guang Zhou, Guangdong, 511400, People's Republic of China
| | - Yu Zou
- Department of Otorhinolaryngology, Guangdong Women and Children Hospital, Guang Zhou, Guangdong, 511400, People's Republic of China
| | - Chun Xie
- Department of Stomatology, People's Hospital of Longhua, Shenzhen 518109, Guangdong, People's Republic of China
| |
Collapse
|
14
|
Benzydamine inhibits osteoclast differentiation and bone resorption via down-regulation of interleukin-1 β expression. Acta Pharm Sin B 2020; 10:462-474. [PMID: 32140392 PMCID: PMC7049613 DOI: 10.1016/j.apsb.2019.11.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 09/23/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022] Open
Abstract
Bone diseases such as osteoporosis and periodontitis are induced by excessive osteoclastic activity, which is closely associated with inflammation. Benzydamine (BA) has been used as a cytokine-suppressive or non-steroidal anti-inflammatory drug that inhibits the production of pro-inflammatory cytokines or prostaglandins. However, its role in osteoclast differentiation and function remains unknown. Here, we explored the role of BA in regulating osteoclast differentiation and elucidated the underlying mechanism. BA inhibited osteoclast differentiation and strongly suppressed interleukin-1β (IL-1β) production. BA inhibited osteoclast formation and bone resorption when added to bone marrow-derived macrophages and differentiated osteoclasts, and the inhibitory effect was reversed by IL-1β treatment. The reporter assay and the inhibitor study of IL-1β transcription suggested that BA inhibited nuclear factor-κB and activator protein-1 by regulating IκB kinase, extracellular signal regulated kinase and P38, resulting in the down-regulation of IL-1β expression. BA also promoted osteoblast differentiation. Furthermore, BA protected lipopolysaccharide- and ovariectomy-induced bone loss in mice, suggesting therapeutic potential against inflammation-induced bone diseases and postmenopausal osteoporosis.
Collapse
|
15
|
Park KH, Gu DR, Kim MS, Lee SH. Inhibitory Effect of Rosae Multiflorae Fructus Extracts on the Receptor Activator of NF-κB Ligand-Induced Osteoclastogenesis through Modulation of P38- and Ca 2+-Mediated Nuclear Factor of Activated T-Cells Cytoplasmic 1 Expression. J Bone Metab 2020; 27:53-63. [PMID: 32190609 PMCID: PMC7064362 DOI: 10.11005/jbm.2020.27.1.53] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 01/31/2020] [Accepted: 02/05/2020] [Indexed: 11/13/2022] Open
Abstract
Background Rosae Multiflorae fructus (RMF), known to have anti-inflammatory and antioxidant properties, has been used as a traditional remedy for inflammatory diseases such as arthritis in Eastern Asia. However, its effect on osteoclasts, which play a crucial role in resorptive inflammatory bone diseases, is yet to be elucidated. Methods The effect of extract of RMF (RMF-E) on receptor activator of nuclear factor-κB ligand (RANKL)-mediated osteoclastogenesis was examined by tartrate-resistant acid phosphatase (TRAP) staining, real-time polymerase chain reaction and western blot analysis. In addition, RANKL-induced Ca2+-oscillation was also investigated. Results RMF-E remarkably inhibited TRAP+-osteoclast and resorptive pit formation in a dose-dependent manner. In addition, the expression of c-Fos and nuclear factor of activated T-cells cytoplasmic, known as pivotal transcription factors for osteoclast formation in vitro and in vivo, and that of the osteoclast differentiation markers such as Acp5, Oscar, CtsK, Atp6v0d2, Tm7sf4, and Nfatc1 were significantly decreased by RMF-E treatment during osteoclastogenesis. The inhibitory effect of RMF-E on RANKL-induced osteoclastogenesis was caused by the suppression of p38 mitogen-activated protein kinase activation, and RANKL-induced Ca2+-oscillation removal via inactivation of Bruton's tyrosine kinase (BTK), and subsequently phospholipase C-γ2. Conclusions RMF-E negatively regulates osteoclast differentiation and formation. These findings suggest the possibility of RMF-E as a traditional therapeutic agent against osteoclast-related bone disorders such as osteoporosis, rheumatoid arthritis, and periodontitis.
Collapse
Affiliation(s)
- Keun Ha Park
- Department of Oral Microbiology and Immunology, College of Dentistry, Wonkwang University, Iksan, Korea
| | - Dong Ryun Gu
- Department of Oral Microbiology and Immunology, College of Dentistry, Wonkwang University, Iksan, Korea
| | - Min Seuk Kim
- Department of Oral Physiology, College of Dentistry, Wonkwang University, Iksan, Korea.,Institute of Biomaterials and Implant, College of Dentistry, Wonkwang University, Iksan, Korea
| | - Seoung Hoon Lee
- Department of Oral Microbiology and Immunology, College of Dentistry, Wonkwang University, Iksan, Korea.,Institute of Biomaterials and Implant, College of Dentistry, Wonkwang University, Iksan, Korea
| |
Collapse
|
16
|
Ihn HJ, Kim TH, Kim K, Kim GY, Jeon YJ, Choi YH, Bae JS, Kim JE, Park EK. 2-O-digalloyl-1,3,4,6-tetra-O-galloyl-β-D-glucose isolated from Galla Rhois suppresses osteoclast differentiation and function by inhibiting NF-κB signaling. BMB Rep 2020. [PMID: 31068248 PMCID: PMC6605525 DOI: 10.5483/bmbrep.2019.52.6.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Natural compounds isolated from medicinal herbs and plants have immense significance in maintaining bone health. Hydrolysable tannins have been shown to possess a variety of medicinal properties including antiviral, anticancer, and anti-osteoclastogenic activities. As a part of a study on the discovery of alternative agent against skeletal diseases, we isolated a hydrolysable tannin, 2-O-digalloyl-1,3,4,6-tetra-O-galloyl- β-D-glucose (DTOGG), from Galla Rhois and examined the effect on osteoclast formation and function. We found that DTOGG significantly inhibited receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation by downregulating the expression of the key regulator in osteoclastogenesis as well as osteoclast-related genes. Analysis of RANKL/RANK signaling revealed that DTOGG impaired activation of IκBα and p65 in the nuclear factor kappa-light-chain- enhancer of activated B cells (NF-κB) signaling pathway. Furthermore, DTOGG reduced bone resorbing activity of osteoclasts, compared to the vehicle-treated control. These results suggest that DTOGG could be a useful natural compound to manage osteoclast-mediated skeletal diseases.
Collapse
Affiliation(s)
- Hye Jung Ihn
- Institute for Hard Tissue and Bio-tooth Regeneration (IHBR), Kyungpook National University, Daegu 41940, Korea
| | - Tae Hoon Kim
- Department of Food Science and Biotechnology, Daegu University, Gyeongsan 38453, Korea
| | - Kiryeong Kim
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, IHBR, Kyungpook National University, Daegu 41940, Korea
| | - Gi-Young Kim
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea
| | - You-Jin Jeon
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, IHBR, Kyungpook National University, Daegu 41940, Korea
| |
Collapse
|