1
|
Zhang H, Li Z, Zhang Z, Li H, Yao Z, Zhang H, Zhao C, Bai X, Pan C, Cai D, Zeng C. Carpaine ameliorates synovial inflammation by promoting p65 degradation and inhibiting the NF-κB signalling pathway. Bone Joint Res 2025; 14:356-367. [PMID: 40237708 PMCID: PMC12002088 DOI: 10.1302/2046-3758.144.bjr-2024-0327.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Aims Osteoarthritis (OA) is a chronic and debilitating joint disease. Despite its prevalence, especially in ageing and obese populations, effective treatments targeting the molecular mechanisms of OA are limited. This study aimed to investigate the role of carpaine (CP), a major alkaloid from the Carica papaya leaf, in inhibiting articular cartilage destruction and synovitis during OA progression, and to elucidate the underlying molecular mechanisms. Methods CP (purity > 98%) was dissolved in dimethyl sulfoxide (DMSO). Various antibodies and reagents were sourced from Sigma-Aldrich, Abcam, and other suppliers. Peritoneal macrophages (pMACs) were cultured in Dulbecco's Modified Eagle Medium (DMEM) and treated with CP to assess its effects on inflammatory cytokine production and nuclear factor-kappa B (NF-κB) signalling. A total of 40 ten-week-old male C57/BL6 mice underwent destabilization of the medial meniscus (DMM) surgery to induce OA. Post-surgery, mice were treated with CP (0.5 or 3 mg/kg) or vehicle via intra-articular injections for up to ten weeks. Cartilage degradation and synovitis were evaluated using Safranin O, Fast Green staining, haematoxylin and eosin (H&E) staining, immunohistochemistry, and quantitative polymerase chain reaction (PCR). Results CP treatment significantly reduced cartilage degeneration and maintained hyaline cartilage thickness compared to the vehicle group. Indicators of cartilage degeneration, such as collagen X (Col X) and matrix metallopeptidase 13 (MMP13), were markedly decreased in the CP-treated group. CP-treated mice exhibited significantly lower synovitis scores at both five and ten weeks post-DMM surgery. CP prominently decreased the production of proinflammatory cytokines (interleukin (IL)-1β, IL-6) in M1 polarized macrophages both in vitro and in vivo. CP impeded NF-κB signalling by promoting p65 degradation through the E3 ubiquitin ligase LRSAM1. The defensive effect of CP was reversed by Lrsam1 small interfering RNA (siRNA), confirming the role of LRSAM1 in CP-mediated NF-κB inhibition. Conclusion CP acts as a 'physiological brake' on NF-κB activation, thereby mitigating synovial inflammation and cartilage destruction in OA. These findings suggest that targeting synovitis via CP could be a promising therapeutic strategy for OA.
Collapse
Affiliation(s)
- Hongbo Zhang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ziyang Li
- Department of Joint Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhicheng Zhang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haobin Li
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zihao Yao
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haiyan Zhang
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Chang Zhao
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiaochun Bai
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chenglong Pan
- Department of Joint Surgery, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Daozhang Cai
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Chun Zeng
- Department of Joint Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Squiers GT, Wan C, Gorder J, Puscher H, Shen J. A Commander-independent function of COMMD3 in endosomal trafficking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.12.628173. [PMID: 39763841 PMCID: PMC11702528 DOI: 10.1101/2024.12.12.628173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Endosomal recycling is a branch of intracellular membrane trafficking that retrieves endocytosed cargo proteins from early and late endosomes to prevent their degradation in lysosomes. A key player in endosomal recycling is the Commander complex, a 16-subunit protein assembly that cooperates with other endosomal factors to recruit cargo proteins and facilitate the formation of tubulo-vesicular carriers. While the crucial role of Commander in endosomal recycling is well established, its molecular mechanism remains poorly understood. Here, we genetically dissected the Commander complex using unbiased genetic screens and comparative targeted mutations. Unexpectedly, our findings revealed a Commander-independent function for COMMD3, a subunit of the Commander complex, in endosomal recycling. COMMD3 regulates a subset of cargo proteins independently of the other Commander subunits. The Commander-independent function of COMMD3 is mediated by its N-terminal domain (NTD), which binds and stabilizes ADP-ribosylation factor 1 (ARF1), a small GTPase regulating endosomal recycling. Mutations disrupting the COMMD3-ARF1 interaction diminish ARF1 expression and impair COMMD3-dependent cargo recycling. These data provide direct evidence that Commander subunits can function outside the holo-complex and raise the intriguing possibility that components of other membrane trafficking complexes may also possess functions beyond their respective complexes.
Collapse
Affiliation(s)
- Galen T. Squiers
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - James Gorder
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Harrison Puscher
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Jingshi Shen
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| |
Collapse
|
3
|
Li S, Zhu H, Zhai Q, Hou Y, Yang Y, Lan H, Jiang M, Xuan J. Exploring Mechanisms of Lang Qing Ata in Non-Alcoholic Steatohepatitis Based on Metabolomics, Network Pharmacological Analysis, and Experimental Validation. Drug Des Devel Ther 2025; 19:1681-1701. [PMID: 40098906 PMCID: PMC11911237 DOI: 10.2147/dddt.s503757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/28/2025] [Indexed: 03/19/2025] Open
Abstract
Background Non-alcoholic steatohepatitis (NASH), as a progressive form of Non-alcoholic fatty liver disease (NAFLD), poses a significant threat to human health as a prevalent and common condition, with a lack of safe and effective therapeutic options. However, the therapeutic effects and potential mechanisms of Lang Qing Ata (LQAtta) against NASH remain elusive. Materials and Methods The components of LQAtta were identified using Ultra-High Performance Liquid Chromatography-Tandem Mass Spectrometry (UHPLC-MS/MS). Subsequently, we employed network construction and analysis approaches within the field of network pharmacology. By integrating known databases and target prediction algorithms, which encompassed database-based target prediction, protein-protein interaction networks, as well as Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses, we unveiled the potential key targets and signaling pathways that these bioactive components might engage with. These discoveries were further validated in subsequent mouse models. An HFHC-induced NASH mouse model was used to validate the therapeutic effects and potential mechanisms of LQAtta on NASH. Results From the UHPLC-MS/MS analysis of LQAtta, a total of 1518 chemical components were identified, with 106 of them being absorbed into the bloodstream. Additionally, based on the acquisition of targets from both LQAtta and the NASH database, a total of 160 common targets were screened. KEGG enrichment analysis indicated that LQAtta may alleviate NASH by modulating pathways such as the Toll-like receptor signaling pathway, the NF-κB signaling pathway, and inflammation-related pathways. In vivo experimental results demonstrated that LQAtta could alleviate liver injury, steatosis, and inflammation induced by NASH, thereby slowing down the disease process. Additionally, LQAtta inhibited the expression and phosphorylation of NF-κB protein, playing a role in preventing NASH. Conclusion In this study, the combination of mass spectrometry analysis, network pharmacology, and animal experiments preliminarily elucidated the potential of LQAtta to treat NASH through NF-κB pathways.
Collapse
Affiliation(s)
- Shupei Li
- Department of Gastroenterology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Hanlong Zhu
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Qi Zhai
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Yu Hou
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Ya Yang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Haifeng Lan
- Department of Gastroenterology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, People’s Republic of China
| | - Mingzuo Jiang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Ji Xuan
- Department of Gastroenterology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
- Department of Gastroenterology and Hepatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
4
|
Gaspa G, Cesarani A, Pauciullo A, Peana I, Macciotta NPP. Genomic Analysis of Sarda Sheep Raised at Diverse Temperatures Highlights Several Genes Involved in Adaptations to the Environment and Heat Stress Response. Animals (Basel) 2024; 14:3585. [PMID: 39765489 PMCID: PMC11672698 DOI: 10.3390/ani14243585] [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: 11/04/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Livestock expresses complex traits influenced by several factors. The response of animals to variations in climatic factors, such as increases in temperature, may induce heat stress conditions. In this study, animals living at different temperatures were compared using the genome-wide Wright fixation index (FST). A total of 825 genotypes of Sarda breed ewes were divided into two groups based on the flocks' average temperature over a 20-year period to compute the FST: 395 and 430 sheep were represented in colder and hotter groups, respectively. After LOWESS regression and CONTROL CHART application, 623 significant markers and 97 selection signatures were found. A total of 280 positional candidate genes were retrieved from a public database. Among these genomic regions, we found 51 annotated genes previously associated with heat stress/tolerance in ruminants (FCGR1A, MDH1, UGP2, MYO1G, and HSPB3), as well as immune response and cellular mechanisms related to how animals cope with thermal stress (RIPK1, SERPINB1, SERPINB9, and PELI1). Moreover, other genes were associated with milk fat (SCD, HERC3, SCFD2, and CHUK), body weight, body fat, and intramuscular fat composition (AGPAT2, ABCD2, MFAP32, YTHDC1, SIRT3, SCD, and RNF121), which might suggest the influence of environmental conditions on the genome of Sarda sheep.
Collapse
Affiliation(s)
- Giustino Gaspa
- Department of Agricultural, Forest and Food Science, University of Torino, 10124 Torino, Italy;
| | - Alberto Cesarani
- Department of Agriculture, University of Sassari, 07100 Sassari, Italy; (A.C.); (N.P.P.M.)
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| | - Alfredo Pauciullo
- Department of Agricultural, Forest and Food Science, University of Torino, 10124 Torino, Italy;
| | - Ilaria Peana
- Servizio Agrometeorologico Regionale per la Sardegna (ARPAS), 07100 Sassari, Italy;
| | - Nicolò P. P. Macciotta
- Department of Agriculture, University of Sassari, 07100 Sassari, Italy; (A.C.); (N.P.P.M.)
| |
Collapse
|
5
|
Almalki WH, Almujri SS. The impact of NF-κB on inflammatory and angiogenic processes in age-related macular degeneration. Exp Eye Res 2024; 248:110111. [PMID: 39326776 DOI: 10.1016/j.exer.2024.110111] [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: 06/16/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Age-related macular degeneration (AMD) is a prominent cause of vision loss, characterized by two different types, dry (atrophic) and wet (neovascular). Dry AMD is distinguished by the progressive deterioration of retinal cells, which ultimately causes a decline in vision. In contrast, wet AMD is defined by the abnormal development of blood vessels underneath the retina, leading to a sudden and severe vision impairment. The course of AMD is primarily driven by chronic inflammation and pathological angiogenesis, in which the NF-κB signaling pathway plays a crucial role. The activation of NF-κB results in the generation of pro-inflammatory cytokines, chemokines, and angiogenic factors like VEGF, which contribute to inflammation and the formation of new blood vessels in AMD. This review analyzes the intricate relationship between NF-κB signaling, inflammation, and angiogenesis in AMD and assesses the possibility of using NF-κB as a target for therapy. The evaluation involves a comprehensive examination of preclinical and clinical evidence that substantiates the effectiveness of NF-κB inhibitors in treating AMD by diminishing inflammation and pathological angiogenesis.
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Aseer, Saudi Arabia
| |
Collapse
|
6
|
Lu J, Zhang J, Jiang H, Hu Z, Zhang Y, He L, Yang J, Xie Y, Wu D, Li H, Zeng K, Tan P, Xiao Q, Song Z, Pan C, Bai X, Yu X. Vangl2 suppresses NF-κB signaling and ameliorates sepsis by targeting p65 for NDP52-mediated autophagic degradation. eLife 2024; 12:RP87935. [PMID: 39269442 PMCID: PMC11398866 DOI: 10.7554/elife.87935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024] Open
Abstract
Van Gogh-like 2 (Vangl2), a core planar cell polarity component, plays an important role in polarized cellular and tissue morphology induction, growth development, and cancer. However, its role in regulating inflammatory responses remains elusive. Here, we report that Vangl2 is upregulated in patients with sepsis and identify Vangl2 as a negative regulator of The nuclear factor-kappaB (NF-κB) signaling by regulating the protein stability and activation of the core transcription component p65. Mice with myeloid-specific deletion of Vangl2 (Vangl2ΔM) are hypersusceptible to lipopolysaccharide (LPS)-induced septic shock. Vangl2-deficient myeloid cells exhibit enhanced phosphorylation and expression of p65, therefore, promoting the secretion of proinflammatory cytokines after LPS stimulation. Mechanistically, NF-κB signaling-induced-Vangl2 recruits E3 ubiquitin ligase PDLIM2 to catalyze K63-linked ubiquitination on p65, which serves as a recognition signal for cargo receptor NDP52-mediated selective autophagic degradation. Taken together, these findings demonstrate Vangl2 as a suppressor of NF-κB-mediated inflammation and provide insights into the crosstalk between autophagy and inflammatory diseases.
Collapse
Affiliation(s)
- Jiansen Lu
- Department of Joint Surgery, the Fifth Affiliated Hospital, Southern Medical UniversityGuangzhouChina
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Jiahuan Zhang
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Huaji Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Department of Orthopaedics, Yuebei People's Hospital Affiliated to Medical College of Shantou UniversityShaoguanChina
| | - Zhiqiang Hu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yufen Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Lian He
- Department of Pharmacology, School of Medicine, Southern University of Science and TechnologyShenzhenChina
- Institute of Biosciences and Technology, College of Medicine, Texas A&M UniversityHoustonUnited States
| | - Jianwu Yang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yingchao Xie
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Dan Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Hongyu Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Ke Zeng
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Peng Tan
- Department of Pharmacology, School of Medicine, Southern University of Science and TechnologyShenzhenChina
- Klarman Cell Observatory, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Qingyue Xiao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Zijing Song
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Chenglong Pan
- Department of Joint Surgery, the Fifth Affiliated Hospital, Southern Medical UniversityGuangzhouChina
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Xiao Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Lab of Single Cell Technology and Application, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
7
|
Fang Q, Xue Y, Yao T, Liu X, Chen J, Han Q, Wang X. Identification of COMMD gene family in large yellow croaker (Larimichthys crocea): Immune response induced by Pseudomonas plecoglossicida infection and acute hypoxia stress. FISH & SHELLFISH IMMUNOLOGY 2024; 152:109780. [PMID: 39033968 DOI: 10.1016/j.fsi.2024.109780] [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: 04/21/2024] [Revised: 07/04/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
The COMMD (Copper Metabolism gene MURR1 Domain) gene family consists of 10 members, which are involved in various biological processes such as copper and sodium transport, NF-κB activity and cell cycle progression. However, the study of COMMD gene family in large yellow croaker (Larimichthys crocea) is largely unknown. In this study, 10 COMMD gene family members (named LcCOMMDs) were successfully identified from large yellow croaker. The results showed that there were differences in the number of LcCOMMDs exons at the level of gene structure, which reflected that they had adjusted and changed accordingly in the process of evolution to adapt to the environment and achieved functional diversification. Through phylogenetic analysis, we found that the LcCOMMDs was highly conserved, indicating their important functions in organisms. It was worth noting that the expression levels of LcCOMMD1, LcCOMMD2, LcCOMMD3, LcCOMMD5 and LcCOMMD10 in the spleen changed significantly after bacterial stress, which suggested that these genes might be involved in the regulation of innate immune response. In addition, the expression levels of LcCOMMD1, LcCOMMD2, LcCOMMD3, LcCOMMD5, LcCOMMD7, LcCOMMD8, LcCOMMD9 and LcCOMMD10 changed significantly after hypoxia exposure, which further proved the role of LcCOMMDs in immune function. In summary, this study not only revealed the important role of COMMD genes in the innate immune response of large yellow croaker, but also provided valuable information for further understanding the regulatory mechanism of COMMD gene family under different conditions.
Collapse
Affiliation(s)
- Qian Fang
- Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, Ningbo, Zhejiang, China.
| | - Yadong Xue
- Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, Ningbo, Zhejiang, China.
| | - TingYan Yao
- Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, Ningbo, Zhejiang, China.
| | - Xiumei Liu
- College of Life Sciences, Yantai University, Yantai, China.
| | - Jianming Chen
- Key Laboratory of Healthy Freshwater Aquaculture, Ministry of Agriculture and Rural Affairs, Key Laboratory of Fish Health and Nutrition of Zhejiang Province, Zhejiang Institute of Freshwater Fisheries, Huzhou, China.
| | - Qingxi Han
- Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, Ningbo, Zhejiang, China.
| | - Xubo Wang
- Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, Ningbo, Zhejiang, China; National Engineering Research Laboratory of Marine Biotechnology and Engineering, Ningbo University, Ningbo, Zhejiang, China; Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, Zhejiang, China; Key Laboratory of Green Mariculture (Co-construction By Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, China.
| |
Collapse
|
8
|
Gromadzka G, Czerwińska J, Krzemińska E, Przybyłkowski A, Litwin T. Wilson's Disease-Crossroads of Genetics, Inflammation and Immunity/Autoimmunity: Clinical and Molecular Issues. Int J Mol Sci 2024; 25:9034. [PMID: 39201720 PMCID: PMC11354778 DOI: 10.3390/ijms25169034] [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: 07/19/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
Wilson's disease (WD) is a rare, autosomal recessive disorder of copper metabolism caused by pathogenic mutations in the ATP7B gene. Cellular copper overload is associated with impaired iron metabolism. Oxidative stress, cuproptosis, and ferroptosis are involved in cell death in WD. The clinical picture of WD is variable. Hepatic/neuropsychiatric/other symptoms may manifest in childhood/adulthood and even old age. It has been shown that phenotypic variability may be determined by the type of ATP7B genetic variants as well as the influence of various genetic/epigenetic, environmental, and lifestyle modifiers. In 1976, immunological abnormalities were first described in patients with WD. These included an increase in IgG and IgM levels and a decrease in the percentage of T lymphocytes, as well as a weakening of their bactericidal effect. Over the following years, it was shown that there is a bidirectional relationship between copper and inflammation. Changes in serum cytokine concentrations and the relationship between cytokine gene variants and the clinical course of the disease have been described in WD patients, as well as in animal models of this disease. Data have also been published on the occurrence of antinuclear antibodies (ANAs), antineutrophil cytoplasmic antibodies (ANCAs), anti-muscle-specific tyrosine kinase antibodies, and anti-acetylcholine receptor antibodies, as well as various autoimmune diseases, including systemic lupus erythematosus (SLE), myasthenic syndrome, ulcerative colitis, multiple sclerosis (MS), polyarthritis, and psoriasis after treatment with d-penicillamine (DPA). The occurrence of autoantibodies was also described, the presence of which was not related to the type of treatment or the form of the disease (hepatic vs. neuropsychiatric). The mechanisms responsible for the occurrence of autoantibodies in patients with WD are not known. It has also not been clarified whether they have clinical significance. In some patients, WD was differentiated or coexisted with an autoimmune disease, including autoimmune hepatitis or multiple sclerosis. Various molecular mechanisms may be responsible for immunological abnormalities and/or the inflammatory processes in WD. Their better understanding may be important for explaining the reasons for the diversity of symptoms and the varied course and response to therapy, as well as for the development of new treatment regimens for WD.
Collapse
Affiliation(s)
- Grażyna Gromadzka
- Department of Biomedical Sciences, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University, Wóycickiego Street 1/3, 01-938 Warsaw, Poland
| | - Julia Czerwińska
- Students Scientific Association “Immunis”, Cardinal Stefan Wyszynski University, Dewajtis Street 5, 01-815 Warsaw, Poland
| | - Elżbieta Krzemińska
- Students Scientific Association “Immunis”, Cardinal Stefan Wyszynski University, Dewajtis Street 5, 01-815 Warsaw, Poland
| | - Adam Przybyłkowski
- Department of Gastroenterology and Internal Medicine, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland;
| | - Tomasz Litwin
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego Street 9, 02-957 Warsaw, Poland;
| |
Collapse
|
9
|
Chen Y, Zhang W, Chen X, Zhang T, Wei H, Huang J, Fan C, Cai M, Wang Y, Zhang Z. Identification, diversity, and evolution analysis of Commd gene family in Haliotis discus hannai and immune response to biotic and abiotic stresses. FISH & SHELLFISH IMMUNOLOGY 2024; 149:109533. [PMID: 38575039 DOI: 10.1016/j.fsi.2024.109533] [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: 01/24/2024] [Revised: 03/30/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
The Commd (Copper Metabolism gene MURR1 Domain) family genes play crucial roles in various biological processes, including copper and sodium transport regulation, NF-κB activity, and cell cycle progression. Their function in Haliotis discus hannai, however, remains unclear. This study focused on identifying and analyzing the Commd genes in H. discus hannai, including their gene structure, phylogenetic relationships, expression profiles, sequence diversity, and alternative splicing. The results revealed significant homology between H. discus hannai's Commd genes and those of other mollusks. Both transcriptome quantitative analysis and qRT-PCR demonstrated the responsiveness of these genes to heat stress and Vibrio parahaemolyticus infection. Notably, alternative splicing analysis revealed that COMMD2, COMMD4, COMMD5, and COMMD7 produce multiple alternative splice variants. Furthermore, sequence diversity analysis uncovered numerous missense mutations, specifically 9 in COMMD5 and 14 in COMMD10. These findings contribute to expanding knowledge on the function and evolution of the Commd gene family and underscore the potential role of COMMD in the innate immune response of H. discus hannai. This research, therefore, offers a novel perspective on the molecular mechanisms underpinning the involvement of Commd genes in innate immunity, paving the way for further explorations in this field.
Collapse
Affiliation(s)
- Yuping Chen
- State Key Laboratory of Mariculture Breeding, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Wenxin Zhang
- State Key Laboratory of Mariculture Breeding, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xin Chen
- State Key Laboratory of Mariculture Breeding, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Tao Zhang
- State Key Laboratory of Mariculture Breeding, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Huina Wei
- State Key Laboratory of Mariculture Breeding, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Jianpeng Huang
- State Key Laboratory of Mariculture Breeding, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Chao Fan
- State Key Laboratory of Mariculture Breeding, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Mingyi Cai
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China; Fujian Engineering Research Center of Aquatic Breeding and Healthy Aquaculture, Xiamen, 361021, China
| | - Yilei Wang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China; Fujian Engineering Research Center of Aquatic Breeding and Healthy Aquaculture, Xiamen, 361021, China.
| | - Ziping Zhang
- State Key Laboratory of Mariculture Breeding, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
10
|
Lin Z, Roche ME, Díaz-Barros V, Domingo-Vidal M, Whitaker-Menezes D, Tuluc M, Uppal G, Caro J, Curry JM, Martinez-Outschoorn U. MiR-200c reprograms fibroblasts to recapitulate the phenotype of CAFs in breast cancer progression. Cell Stress 2024; 8:1-20. [PMID: 38476765 PMCID: PMC10927306 DOI: 10.15698/cst2024.03.293] [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: 06/15/2023] [Revised: 12/20/2023] [Accepted: 01/11/2024] [Indexed: 03/14/2024] Open
Abstract
Mesenchymal-epithelial plasticity driving cancer progression in cancer-associated fibroblasts (CAFs) is undetermined. This work identifies a subgroup of CAFs in human breast cancer exhibiting mesenchymal-to-epithelial transition (MET) or epithelial-like profile with high miR-200c expression. MiR-200c overexpression in fibroblasts is sufficient to drive breast cancer aggressiveness. Oxidative stress in the tumor microenvironment induces miR-200c by DNA demethylation. Proteomics, RNA-seq and functional analyses reveal that miR-200c is a novel positive regulator of NFκB-HIF signaling via COMMD1 downregulation and stimulates pro-tumorigenic inflammation and glycolysis. Reprogramming fibroblasts toward MET via miR-200c reduces stemness and induces a senescent phenotype. This pro-tumorigenic profile in CAFs fosters carcinoma cell resistance to apoptosis, proliferation and immunosuppression, leading to primary tumor growth, metastases, and resistance to immuno-chemotherapy. Conversely, miR-200c inhibition in fibroblasts restrains tumor growth with abated oxidative stress and an anti-tumorigenic immune environment. This work determines the mechanisms by which MET in CAFs via miR-200c transcriptional enrichment with DNA demethylation triggered by oxidative stress promotes cancer progression. CAFs undergoing MET trans-differentiation and senescence coordinate heterotypic signaling that may be targeted as an anti-cancer strategy.
Collapse
Affiliation(s)
- Zhao Lin
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Megan E. Roche
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Víctor Díaz-Barros
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Marina Domingo-Vidal
- Immunology, Microenvironment & Metastasis Program, Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Diana Whitaker-Menezes
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Madalina Tuluc
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Guldeep Uppal
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jaime Caro
- Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Joseph M. Curry
- Department of Otolaryngology-Head and Neck Surgery, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ubaldo Martinez-Outschoorn
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
11
|
Klaus T, Hieber C, Bros M, Grabbe S. Integrins in Health and Disease-Suitable Targets for Treatment? Cells 2024; 13:212. [PMID: 38334604 PMCID: PMC10854705 DOI: 10.3390/cells13030212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/13/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
Integrin receptors are heterodimeric surface receptors that play multiple roles regarding cell-cell communication, signaling, and migration. The four members of the β2 integrin subfamily are composed of an alternative α (CD11a-d) subunit, which determines the specific receptor properties, and a constant β (CD18) subunit. This review aims to present insight into the multiple immunological roles of integrin receptors, with a focus on β2 integrins that are specifically expressed by leukocytes. The pathophysiological role of β2 integrins is confirmed by the drastic phenotype of patients suffering from leukocyte adhesion deficiencies, most often resulting in severe recurrent infections and, at the same time, a predisposition for autoimmune diseases. So far, studies on the role of β2 integrins in vivo employed mice with a constitutive knockout of all β2 integrins or either family member, respectively, which complicated the differentiation between the direct and indirect effects of β2 integrin deficiency for distinct cell types. The recent generation and characterization of transgenic mice with a cell-type-specific knockdown of β2 integrins by our group has enabled the dissection of cell-specific roles of β2 integrins. Further, integrin receptors have been recognized as target receptors for the treatment of inflammatory diseases as well as tumor therapy. However, whereas both agonistic and antagonistic agents yielded beneficial effects in animal models, the success of clinical trials was limited in most cases and was associated with unwanted side effects. This unfavorable outcome is most probably related to the systemic effects of the used compounds on all leukocytes, thereby emphasizing the need to develop formulations that target distinct types of leukocytes to modulate β2 integrin activity for therapeutic applications.
Collapse
Affiliation(s)
| | | | | | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (T.K.); (C.H.); (M.B.)
| |
Collapse
|
12
|
Lee CM, Go YY, Song JJ. Inhibition of lipopolysaccharide-induced inflammation by trophoblast-conditioned medium and trophoblast-derived extracellular vesicles in human middle ear epithelial cells. Sci Rep 2023; 13:19822. [PMID: 37963902 PMCID: PMC10645728 DOI: 10.1038/s41598-023-46731-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 11/04/2023] [Indexed: 11/16/2023] Open
Abstract
Otitis media is a common disease but can cause severe inner ear inflammation and hearing loss if it persists for more than two weeks. This study elucidates the inflammation-inhibiting efficacy of conditioned medium (CM) and extracellular vesicles (EVs) derived from human trophoblast (TB) cells in lipopolysaccharide (LPS)-induced human middle ear epithelial cells (HMEECs). TB-conditioned medium (TB-CM) reduced the inflammatory response and regulated mucin and epithelial sodium channel genes in LPS-induced HMEECs. The underlying mechanism of cell migration during inflammatory healing in LPS-induced HMEECs treated with TB-CM was determined by RNA-sequencing analysis. Specifically, the NF-κB pathway related to the copper metabolism MURR1 domain protein was studied and verified through siRNA. This elucidation of the anti-inflammatory effect of TB-CM and TB-derived EVs demonstrates their clinical potential to treat chronic inflammation.
Collapse
Affiliation(s)
- Chan Mi Lee
- Division of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yoon Young Go
- Division of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
- Center for Health Care Convergence at Korea University Guro Hospital, Seoul, Republic of Korea
| | - Jae-Jun Song
- Division of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea.
- Center for Health Care Convergence at Korea University Guro Hospital, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Healy MD, McNally KE, Butkovič R, Chilton M, Kato K, Sacharz J, McConville C, Moody ERR, Shaw S, Planelles-Herrero VJ, Yadav SKN, Ross J, Borucu U, Palmer CS, Chen KE, Croll TI, Hall RJ, Caruana NJ, Ghai R, Nguyen THD, Heesom KJ, Saitoh S, Berger I, Schaffitzel C, Williams TA, Stroud DA, Derivery E, Collins BM, Cullen PJ. Structure of the endosomal Commander complex linked to Ritscher-Schinzel syndrome. Cell 2023; 186:2219-2237.e29. [PMID: 37172566 PMCID: PMC10187114 DOI: 10.1016/j.cell.2023.04.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/23/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023]
Abstract
The Commander complex is required for endosomal recycling of diverse transmembrane cargos and is mutated in Ritscher-Schinzel syndrome. It comprises two sub-assemblies: Retriever composed of VPS35L, VPS26C, and VPS29; and the CCC complex which contains twelve subunits: COMMD1-COMMD10 and the coiled-coil domain-containing (CCDC) proteins CCDC22 and CCDC93. Combining X-ray crystallography, electron cryomicroscopy, and in silico predictions, we have assembled a complete structural model of Commander. Retriever is distantly related to the endosomal Retromer complex but has unique features preventing the shared VPS29 subunit from interacting with Retromer-associated factors. The COMMD proteins form a distinctive hetero-decameric ring stabilized by extensive interactions with CCDC22 and CCDC93. These adopt a coiled-coil structure that connects the CCC and Retriever assemblies and recruits a 16th subunit, DENND10, to form the complete Commander complex. The structure allows mapping of disease-causing mutations and reveals the molecular features required for the function of this evolutionarily conserved trafficking machinery.
Collapse
Affiliation(s)
- Michael D Healy
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Kerrie E McNally
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK; MRC Laboratory of Molecular Biology, CB2 0QH Cambridge, UK.
| | - Rebeka Butkovič
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK
| | - Molly Chilton
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK
| | - Kohji Kato
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK
| | - Joanna Sacharz
- Department of Biochemistry and Pharmacology, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Calum McConville
- Department of Biochemistry and Pharmacology, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Edmund R R Moody
- School of Biological Sciences, University of Bristol, BS8 1TD Bristol, UK
| | - Shrestha Shaw
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK
| | | | - Sathish K N Yadav
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK
| | - Jennifer Ross
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK
| | - Ufuk Borucu
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK
| | - Catherine S Palmer
- Department of Biochemistry and Pharmacology, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Kai-En Chen
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Tristan I Croll
- Cambridge Institute for Medical Research, University of Cambridge, CB2 0XY Cambridge, UK
| | - Ryan J Hall
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Nikeisha J Caruana
- Department of Biochemistry and Pharmacology, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia; Institute of Health and Sport (iHeS), Victoria University, Melbourne, VIC Australia
| | - Rajesh Ghai
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Thi H D Nguyen
- MRC Laboratory of Molecular Biology, CB2 0QH Cambridge, UK
| | - Kate J Heesom
- Proteomics Facility, School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Imre Berger
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK; Max Planck Bristol Centre for Minimal Biology, Department of Chemistry, University of Bristol, BS8 1TS Bristol, UK
| | - Christiane Schaffitzel
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK
| | - Tom A Williams
- School of Biological Sciences, University of Bristol, BS8 1TD Bristol, UK
| | - David A Stroud
- Department of Biochemistry and Pharmacology, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia; Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne, VIC Australia
| | | | - Brett M Collins
- Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia.
| | - Peter J Cullen
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, BS8 1TD Bristol, UK.
| |
Collapse
|
14
|
Kovács P, Pushparaj PN, Takács R, Mobasheri A, Matta C. The clusterin connectome: Emerging players in chondrocyte biology and putative exploratory biomarkers of osteoarthritis. Front Immunol 2023; 14:1103097. [PMID: 37033956 PMCID: PMC10081159 DOI: 10.3389/fimmu.2023.1103097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
IntroductionClusterin is amoonlighting protein that hasmany functions. It is amultifunctional Q6 holdase chaperone glycoprotein that is present intracellularly and extracellularly in almost all bodily fluids. Clusterin is involved in lipid transport, cell differentiation, regulation of apoptosis, and clearance of cellular debris, and plays a protective role in ensuring cellular survival. However, the possible involvement of clusterin in arthritic disease remains unclear. Given the significant potential of clusterin as a biomarker of osteoarthritis (OA), a more detailed analysis of its complex network in an inflammatory environment, specifically in the context of OA, is required. Based on the molecular network of clusterin, this study aimed to identify interacting partners that could be developed into biomarker panels for OA.MethodsThe STRING database and Cytoscape were used to map and visualize the clusterin connectome. The Qiagen Ingenuity Pathway Analysis (IPA) software was used to analyze and study clusterinassociated signaling networks in OA. We also analyzed transcription factors known to modulate clusterin expression, which may be altered in OA.ResultsThe top hits in the clusterin network were intracellular chaperones, aggregate-forming proteins, apoptosis regulators and complement proteins. Using a text-mining approach in Cytoscape, we identified additional interacting partners, including serum proteins, apolipoproteins, and heat shock proteins.DiscussionBased on known interactions with proteins, we predicted potential novel components of the clusterin connectome in OA, including selenoprotein R, semaphorins, and meprins, which may be important for designing new prognostic or diagnostic biomarker panels.
Collapse
Affiliation(s)
- Patrik Kovács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research (CEGMR), Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Roland Takács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ali Mobasheri
- FibroHealth Interdisciplinary Research Programme, Fibrobesity Cluster, Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
- *Correspondence: Csaba Matta, ; Ali Mobasheri,
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- *Correspondence: Csaba Matta, ; Ali Mobasheri,
| |
Collapse
|
15
|
You G, Zhou C, Wang L, Liu Z, Fang H, Yao X, Zhang X. COMMD proteins function and their regulating roles in tumors. Front Oncol 2023; 13:1067234. [PMID: 36776284 PMCID: PMC9910083 DOI: 10.3389/fonc.2023.1067234] [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: 10/11/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
The COMMD proteins are a highly conserved protein family with ten members that play a crucial role in a variety of biological activities, including copper metabolism, endosomal sorting, ion transport, and other processes. Recent research have demonstrated that the COMMD proteins are closely associated with a wide range of disorders, such as hepatitis, myocardial ischemia, cerebral ischemia, HIV infection, and cancer. Among these, the role of COMMD proteins in tumors has been thoroughly explored; they promote or inhibit cancers such as lung cancer, liver cancer, gastric cancer, and prostate cancer. COMMD proteins can influence tumor proliferation, invasion, metastasis, and tumor angiogenesis, which are strongly related to the prognosis of tumors and are possible therapeutic targets for treating tumors. In terms of molecular mechanism, COMMD proteins in tumor cells regulate the oncogenes of NF-κB, HIF, c-MYC, and others, and are related to signaling pathways including apoptosis, autophagy, and ferroptosis. For the clinical diagnosis and therapy of malignancies, additional research into the involvement of COMMD proteins in cancer is beneficial.
Collapse
Affiliation(s)
- Guangqiang You
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Jilin University, Jilin University, Changchun, China
| | - Chen Zhou
- Department of General Affairs, First Hospital of Jilin University (the Eastern Division), Jilin University, Changchun, China
| | - Lei Wang
- Department of Pediatric Neurology, First Hospital of Jilin University, Jilin University, Changchun, China
| | - Zefeng Liu
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Jilin University, Jilin University, Changchun, China
| | - He Fang
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Jilin University, Jilin University, Changchun, China
| | - Xiaoxao Yao
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Jilin University, Jilin University, Changchun, China,*Correspondence: Xiaoxao Yao, ; Xuewen Zhang,
| | - Xuewen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Jilin University, Jilin University, Changchun, China,*Correspondence: Xiaoxao Yao, ; Xuewen Zhang,
| |
Collapse
|
16
|
Medina L, Guerrero-Muñoz J, Castillo C, Liempi A, Fernández-Moya A, Araneda S, Ortega Y, Rivas C, Maya JD, Kemmerling U. Differential microRNAs expression during ex vivo infection of canine and ovine placental explants with Trypanosoma cruzi and Toxoplasma gondii. Acta Trop 2022; 235:106651. [PMID: 35964709 DOI: 10.1016/j.actatropica.2022.106651] [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: 05/27/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022]
Abstract
Trypanosoma cruzi and Toxoplasma gondii are two zoonotic parasites that constitute significant human and animal health threats, causing a significant economic burden worldwide. Both parasites can be transmitted congenitally, but transmission rates for T. gondii are high, contrary to what has been observed for T. cruzi. The probability of congenital transmission depends on complex interactions between the pathogen and the host, including the modulation of host cell gene expression by miRNAs. During ex vivo infection of canine and ovine placental explants, we evaluated the expression of 3 miRNAs (miR-30e-3p, miR-3074-5p, and miR-127-3p) previously associated with parasitic and placental diseases and modulated by both parasites. In addition, we identified the possible target genes of the miRNAs by using computational prediction tools and performed GO and KEGG enrichment analyses to identify the biological functions and associated pathologies. The three miRNAs are differentially expressed in the canine and ovine placenta in response to T. cruzi and T. gondii. We conclude that the observed differential expression and associated functions might explain, at least partially, the differences in transmission rates and susceptibility to parasite infection in different species.
Collapse
Affiliation(s)
- Lisvaneth Medina
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Jesús Guerrero-Muñoz
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Christian Castillo
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Núcleo de Investigación Aplicada en Ciencias Veterinarias y Agronómicas, Facultad de Medicina Veterinaria y Agronomía, Universidad de Las Américas, Chile
| | - Ana Liempi
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alejandro Fernández-Moya
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sebastian Araneda
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Departamento de Patología y Medicina Oral, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Yessica Ortega
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Facultad de Farmacia y Bioanálisis, Universidad de Los Andes, Mérida, Venezuela
| | - Cristian Rivas
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Juan Diego Maya
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ulrike Kemmerling
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
17
|
Bednarczyk M, Bolduan V, Haist M, Stege H, Hieber C, Johann L, Schelmbauer C, Blanfeld M, Karram K, Schunke J, Klaus T, Tubbe I, Montermann E, Röhrig N, Hartmann M, Schlosser J, Bopp T, Clausen BE, Waisman A, Bros M, Grabbe S. β2 Integrins on Dendritic Cells Modulate Cytokine Signaling and Inflammation-Associated Gene Expression, and Are Required for Induction of Autoimmune Encephalomyelitis. Cells 2022; 11:cells11142188. [PMID: 35883631 PMCID: PMC9322999 DOI: 10.3390/cells11142188] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 01/27/2023] Open
Abstract
Heterodimeric β2 integrin surface receptors (CD11a-d/CD18) are specifically expressed by leukocytes that contribute to pathogen uptake, cell migration, immunological synapse formation and cell signaling. In humans, the loss of CD18 expression results in leukocyte adhesion deficiency syndrome (LAD-)1, largely characterized by recurrent severe infections. All available mouse models display the constitutive and ubiquitous knockout of either α or the common β2 (CD18) subunit, which hampers the analysis of the cell type-specific role of β2 integrins in vivo. To overcome this limitation, we generated a CD18 gene floxed mouse strain. Offspring generated from crossing with CD11c-Cre mice displayed the efficient knockdown of β2 integrins, specifically in dendritic cells (DCs). Stimulated β2-integrin-deficient splenic DCs showed enhanced cytokine production and the concomitantly elevated activity of signal transducers and activators of transcription (STAT) 1, 3 and 5, as well as the impaired expression of suppressor of cytokine signaling (SOCS) 2–6 as assessed in bone marrow-derived (BM) DCs. Paradoxically, these BMDCs also showed the attenuated expression of genes involved in inflammatory signaling. In line, in experimental autoimmune encephalomyelitis mice with a conditional DC-specific β2 integrin knockdown presented with a delayed onset and milder course of disease, associated with lower frequencies of T helper cell populations (Th)1/Th17 in the inflamed spinal cord. Altogether, our mouse model may prove to be a valuable tool to study the leukocyte-specific functions of β2 integrins in vivo.
Collapse
Affiliation(s)
- Monika Bednarczyk
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Vanessa Bolduan
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Maximilian Haist
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Henner Stege
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Christoph Hieber
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Lisa Johann
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
| | - Carsten Schelmbauer
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
| | - Michaela Blanfeld
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
| | - Khalad Karram
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Jenny Schunke
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Tanja Klaus
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Ingrid Tubbe
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Evelyn Montermann
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Nadine Röhrig
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Maike Hartmann
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Jana Schlosser
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Tobias Bopp
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
- Institute of Immunology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Matthias Bros
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
- Correspondence: ; Tel.: +49-61-3117-4412
| |
Collapse
|
18
|
Wang B, Shen J. NF-κB Inducing Kinase Regulates Intestinal Immunity and Homeostasis. Front Immunol 2022; 13:895636. [PMID: 35833111 PMCID: PMC9271571 DOI: 10.3389/fimmu.2022.895636] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/31/2022] [Indexed: 11/22/2022] Open
Abstract
Intestinal immunity and homeostasis are maintained through the regulation of cytokine trafficking, microbiota, necrosis and apoptosis. Intestinal immunity and homeostasis participate in host defenses and inflammatory responses locally or systemically through the gut-organ axis. NF-κB functions as a crucial transcription factor mediating the expression of proteins related to the immune responses. The activation of NF-κB involves two major pathways: canonical and non-canonical. The canonical pathway has been extensively studied and reviewed. Here, we present the current knowledge of NIK, a pivotal mediator of the non-canonical NF-κB pathway and its role in intestinal immunity and homeostasis. This review also discusses the novel role of NIK signaling in the pathogenesis and treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Bingran Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
- Ottawa-Shanghai Joint School of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Shanghai Institute of Digestive Disease, Shanghai Jiao Tong University, Shanghai, China
- Ottawa-Shanghai Joint School of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Jun Shen,
| |
Collapse
|
19
|
Díez-Tercero L, Delgado LM, Perez RA. Modulation of Macrophage Response by Copper and Magnesium Ions in Combination with Low Concentrations of Dexamethasone. Biomedicines 2022; 10:biomedicines10040764. [PMID: 35453514 PMCID: PMC9030383 DOI: 10.3390/biomedicines10040764] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/09/2022] [Accepted: 03/22/2022] [Indexed: 12/16/2022] Open
Abstract
Macrophages have been deemed crucial for correct tissue regeneration, which is a complex process with multiple overlapping phases, including inflammation. Previous studies have suggested that divalent ions are promising cues that can induce an anti-inflammatory response, since they are stable cues that can be released from biomaterials. However, their immunomodulatory potential is limited in a pro-inflammatory environment. Therefore, we investigated whether copper and magnesium ions combined with low concentrations of the anti-inflammatory drug, dexamethasone (dex), could have a synergistic effect in macrophage, with or without pro-inflammatory stimulus, in terms of morphology, metabolic activity and gene expression. Our results showed that the combination of copper and dex strongly decreased the expression of pro-inflammatory markers, while the combination with magnesium upregulated the expression of IL-10. Moreover, in the presence of a pro-inflammatory stimulus, the combination of copper and dex induced a strong TNF-α response, suggesting an impairment of the anti-inflammatory actions of dex. The combination of magnesium and dex in the presence of a pro-inflammatory stimulus did not promote any improvement in comparison to dex alone. The results obtained in this study could be relevant for tissue engineering applications and in the design of platforms with a dual release of divalent ions and small molecules.
Collapse
Affiliation(s)
- Leire Díez-Tercero
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain;
- Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain
| | - Luis M. Delgado
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain;
- Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain
- Correspondence: (L.M.D.); (R.A.P.); Tel.: +34-935042000 (L.M.D. & R.A.P.)
| | - Roman A. Perez
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain;
- Basic Science Department, Universitat Internacional de Catalunya, Sant Cugat del Vallès, 08195 Barcelona, Spain
- Correspondence: (L.M.D.); (R.A.P.); Tel.: +34-935042000 (L.M.D. & R.A.P.)
| |
Collapse
|
20
|
Neveu B, Richer C, Cassart P, Caron M, Jimenez-Cortes C, St-Onge P, Fuchs C, Garnier N, Gobeil S, Sinnett D. Identification of new ETV6 modulators through a high-throughput functional screening. iScience 2022; 25:103858. [PMID: 35198911 PMCID: PMC8851229 DOI: 10.1016/j.isci.2022.103858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/01/2022] [Accepted: 01/28/2022] [Indexed: 12/02/2022] Open
Abstract
ETV6 transcriptional activity is critical for proper blood cell development in the bone marrow. Despite the accumulating body of evidence linking ETV6 malfunction to hematological malignancies, its regulatory network remains unclear. To uncover genes that modulate ETV6 repressive transcriptional activity, we performed a specifically designed, unbiased genome-wide shRNA screen in pre-B acute lymphoblastic leukemia cells. Following an extensive validation process, we identified 13 shRNAs inducing overexpression of ETV6 transcriptional target genes. We showed that the silencing of AKIRIN1, COMMD9, DYRK4, JUNB, and SRP72 led to an abrogation of ETV6 repressive activity. We identified critical modulators of the ETV6 function which could participate in cellular transformation through the ETV6 transcriptional network. We develop a genome-wide shRNAs screen for ETV6 modulators The screen uncovered 13 novel putative ETV6 modulator genes The modulators demonstrated a broad impact on the ETV6 transcriptional network T-ALL cells results suggest modulators are conserved in other cellular contexts
Collapse
Affiliation(s)
- Benjamin Neveu
- Sainte-Justine University Health Center Research Center, Montreal, QC H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Chantal Richer
- Sainte-Justine University Health Center Research Center, Montreal, QC H3T 1C5, Canada
| | - Pauline Cassart
- Sainte-Justine University Health Center Research Center, Montreal, QC H3T 1C5, Canada
| | - Maxime Caron
- Sainte-Justine University Health Center Research Center, Montreal, QC H3T 1C5, Canada
- Department of Human Genetics, McGill University, Montréal, QC H3A 0C7, Canada
| | - Camille Jimenez-Cortes
- Sainte-Justine University Health Center Research Center, Montreal, QC H3T 1C5, Canada
- Molecular Biology Program, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Pascal St-Onge
- Sainte-Justine University Health Center Research Center, Montreal, QC H3T 1C5, Canada
| | - Claire Fuchs
- Sainte-Justine University Health Center Research Center, Montreal, QC H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
| | - Nicolas Garnier
- Sainte-Justine University Health Center Research Center, Montreal, QC H3T 1C5, Canada
| | - Stéphane Gobeil
- CHU de Québec-Université Laval Research Center, Quebec City, QC G1V 4G2, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada
- Corresponding author
| | - Daniel Sinnett
- Sainte-Justine University Health Center Research Center, Montreal, QC H3T 1C5, Canada
- Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, QC H3C 3J7, Canada
- Corresponding author
| |
Collapse
|
21
|
Xiao Y, Wu L, He L, Tang Y, Guo S, Zhai S. Transcriptomic analysis using dual RNA sequencing revealed a Pathogen-Host interaction after Edwardsiella anguillarum infection in European eel (Anguilla anguilla). FISH & SHELLFISH IMMUNOLOGY 2022; 120:745-757. [PMID: 34974154 DOI: 10.1016/j.fsi.2021.12.051] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/14/2021] [Accepted: 12/28/2021] [Indexed: 05/26/2023]
Abstract
Many studies have explored differentially expressed genes (DEGs) between some pathogens and hosts, but no study has focused on the interaction of DEGs between Edwardsiella anguillarum (Ea) and Anguilla anguilla (Aa). In this study, we examined the interactions of DEGs during Ea infection and Aa anti-infection processes by dual RNA sequencing. Total RNA from in vitro and in vivo (Aa liver) Ea culture was extracted. Using high-throughput transcriptomics, significant DEGs that were expressed between Ea cultured in vitro versus in vivo and those in the liver of the infected group versus control group were identified. Protein-protein interactions between the pathogen and host were explored using Cytoscape according to the HPIDB 3.0 interaction transcription database. The results showed that the liver in the infection group presented with severe bleeding and a large number of thrombi in the hepatic vessels. We found 490 upregulated and 398 downregulated DEGs of Ea in vivo versus Ea cultured in vitro, and 2177 upregulated and 970 downregulated genes in the liver of the infected eels. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of the pathogen DEGs revealed that the upregulated genes were mainly enriched in migration, colonization, biofilm formation, and significantly enriched in ABC transport and quorum sensing; the downregulated genes were mainly involved in metabolism, information transduction, organelle formation, enzyme catalysis, molecular transport, and binding. GO of the host DEGs showed that metabolic process, catalytic activity, single organism metabolic process, small molecule binding, nucleotide binding, nucleotide phosphate binding, and anion binding were markedly enriched. Finally, we found that 79 Ea and 148 Aa proteins encoded by these DEGs were involved in an interaction network, and some pathogen (DegP, gcvP, infC, carB, rpoC, trpD, sthA, and FhuB) and host proteins (MANBA, STAT1, ETS2, ZEP1, TKT1, NMI and RBPMS) appear to play crucial roles in infection. Thus, determining the interaction networks revealed crucial molecular mechanisms underlying the process of pathogenic infection and host anti-infection.
Collapse
Affiliation(s)
- Yiqun Xiao
- Fisheries College, Jimei University/Engineering Research Center of the Modern Industry Technology for Eel. Ministry of Education of PR China, Xiamen, 361021, China
| | - Liqun Wu
- College of Overseas Education, Jimei University, Xiamen, 361021, China
| | - Le He
- Fisheries College, Jimei University/Engineering Research Center of the Modern Industry Technology for Eel. Ministry of Education of PR China, Xiamen, 361021, China
| | - Yijun Tang
- Department of Chemistry, University of Wisconsin Oshkosh, 800 Algoma Blvd, Oshkosh, WI, USA
| | - Songlin Guo
- Fisheries College, Jimei University/Engineering Research Center of the Modern Industry Technology for Eel. Ministry of Education of PR China, Xiamen, 361021, China.
| | - Shaowei Zhai
- Fisheries College, Jimei University/Engineering Research Center of the Modern Industry Technology for Eel. Ministry of Education of PR China, Xiamen, 361021, China.
| |
Collapse
|
22
|
Cohen K, Mouhadeb O, Ben Shlomo S, Langer M, Neumann A, Erez N, Moshkovits I, Pelet R, Kedar DJ, Brazowski E, Guilliams M, Goodridge HS, Gluck N, Varol C. COMMD10 is critical for Kupffer cell survival and controls Ly6C hi monocyte differentiation and inflammation in the injured liver. Cell Rep 2021; 37:110026. [PMID: 34788631 DOI: 10.1016/j.celrep.2021.110026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 09/27/2021] [Accepted: 10/28/2021] [Indexed: 12/23/2022] Open
Abstract
Liver-resident macrophages Kupffer cells (KCs) and infiltrating Ly6Chi monocytes both contribute to liver tissue regeneration in various pathologies but also to disease progression upon disruption of orderly consecutive regeneration cascades. Little is known about molecular pathways that regulate their differentiation, maintenance, or inflammatory behavior during injury. Here, we show that copper metabolism MURR1 domain (COMMD)10-deficient KCs adopt liver-specific identity. Strikingly, COMMD10 deficiency in KCs and in other tissue-resident macrophages impedes their homeostatic survival, leading to their continuous replacement by Ly6Chi monocytes. While COMMD10 deficiency in KCs mildly worsens acetaminophen-induced liver injury (AILI), its deficiency in Ly6Chi monocytes results in exacerbated and sustained hepatic damage. Monocytes display unleashed inflammasome activation and a reduced type I interferon response and acquire "neutrophil-like" and lipid-associated macrophage differentiation fates. Collectively, COMMD10 appears indispensable for KC and other tissue-resident macrophage survival and is an important regulator of Ly6Chi monocyte fate decisions and reparative behavior in the diseased liver.
Collapse
Affiliation(s)
- Keren Cohen
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel; Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Odelia Mouhadeb
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel; Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Shani Ben Shlomo
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Marva Langer
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel; Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Anat Neumann
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Noam Erez
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Itay Moshkovits
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel; Internal Medicine T, Sourasky Medical Center, Tel-Aviv 64239, Israel
| | - Rotem Pelet
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Daniel J Kedar
- Department of Plastic and Reconstructive Surgery, Sourasky Medical Center, Tel-Aviv 64239, Israel
| | - Eli Brazowski
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel
| | - Martin Guilliams
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Helen S Goodridge
- Board of Governors Regenerative Medicine Institute and Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nathan Gluck
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel.
| | - Chen Varol
- Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center, and Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 64239, Israel; Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel.
| |
Collapse
|
23
|
Heida A, Gruben N, Catrysse L, Koehorst M, Koster M, Kloosterhuis NJ, Gerding A, Havinga R, Bloks VW, Bongiovanni L, Wolters JC, van Dijk T, van Loo G, de Bruin A, Kuipers F, Koonen DPY, van de Sluis B. The hepatocyte IKK:NF-κB axis promotes liver steatosis by stimulating de novo lipogenesis and cholesterol synthesis. Mol Metab 2021; 54:101349. [PMID: 34626855 PMCID: PMC8581577 DOI: 10.1016/j.molmet.2021.101349] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/20/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Obesity-related chronic inflammation plays an important role in the development of Metabolic Associated Fatty Liver Disease (MAFLD). Although the contribution of the pro-inflammatory NF-κB signaling pathway to the progression from simple steatosis to non-alcoholic steatohepatitis (NASH) is well-established, its role as an initiator of hepatic steatosis and the underlying mechanism remains unclear. Here, we investigated the hypothesis that the hepatocytic NF-κB signaling pathway acts as a metabolic regulator, thereby promoting hepatic steatosis development. METHODS A murine model expressing a constitutively active form of IKKβ in hepatocytes (Hep-IKKβca) was used to activate hepatocyte NF-κB. In addition, IKKβca was also expressed in hepatocyte A20-deficient mice (IKKβca;A20LKO). A20 is an NF-κB-target gene that inhibits the activation of the NF-κB signaling pathway upstream of IKKβ. These mouse models were fed a sucrose-rich diet for 8 weeks. Hepatic lipid levels were measured and using [1-13C]-acetate de novo lipogenesis and cholesterol synthesis rate were determined. Gene expression analyses and immunoblotting were used to study the lipogenesis and cholesterol synthesis pathways. RESULTS Hepatocytic NF-κB activation by expressing IKKβca in hepatocytes resulted in hepatic steatosis without inflammation. Ablation of hepatocyte A20 in Hep-IKKβca mice (IKKβca;A20LKO mice) exacerbated hepatic steatosis, characterized by macrovesicular accumulation of triglycerides and cholesterol, and increased plasma cholesterol levels. Both De novo lipogenesis (DNL) and cholesterol synthesis were found elevated in IKKβca;A20LKO mice. Phosphorylation of AMP-activated kinase (AMPK) - a suppressor in lipogenesis and cholesterol synthesis - was decreased in IKKβca;A20LKO mice. This was paralleled by elevated protein levels of hydroxymethylglutaryl-CoA synthase 1 (HMGCS1) and reduced phosphorylation of HMG-CoA reductase (HMGCR) both key enzymes in the cholesterol synthesis pathway. Whereas inflammation was not observed in young IKKβca;A20LKO mice sustained hepatic NF-κB activation resulted in liver inflammation, together with elevated hepatic and plasma cholesterol levels in middle-aged mice. CONCLUSIONS The hepatocytic IKK:NF-κB axis is a metabolic regulator by controlling DNL and cholesterol synthesis, independent of its central role in inflammation. The IKK:NF-κB axis controls the phosphorylation levels of AMPK and HMGCR and the protein levels of HMGCS1. Chronic IKK-mediated NF-κB activation may contribute to the initiation of hepatic steatosis and cardiovascular disease risk in MAFLD patients.
Collapse
Affiliation(s)
- Andries Heida
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nanda Gruben
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Leen Catrysse
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Martijn Koehorst
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mirjam Koster
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Niels J Kloosterhuis
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Albert Gerding
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rick Havinga
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Vincent W Bloks
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Laura Bongiovanni
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Justina C Wolters
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Theo van Dijk
- Departments of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Geert van Loo
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Alain de Bruin
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands
| | - Folkert Kuipers
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Departments of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Debby P Y Koonen
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Bart van de Sluis
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
24
|
Wang X, He S, Zheng X, Huang S, Chen H, Chen H, Luo W, Guo Z, He X, Zhao Q. Transcriptional analysis of the expression, prognostic value and immune infiltration activities of the COMMD protein family in hepatocellular carcinoma. BMC Cancer 2021; 21:1001. [PMID: 34493238 PMCID: PMC8424899 DOI: 10.1186/s12885-021-08699-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 08/17/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The copper metabolism MURR1 domain (COMMD) protein family involved in tumor development and progression in several types of human cancer, but little is known about the function of COMMD proteins in hepatocellular carcinoma (HCC). METHODS The ONCOMINE and the UALCAN databases were used to evaluate the expression of COMMD1-10 in HCC and the association of this family with individual cancer stage and tumor grade. Kaplan-Meier (K-M) Plotter and Cox analysis hint the prognostic value of COMMDs. A network comprising 50 most similar genes and COMMD1-10 was constructed with the STRING database. Gene set enrichment analysis (GSEA) was performed using LinkedOmics database. The correlations between COMMD expression and the presence of immune infiltrating cells were also analyzed by the tumor immune estimation resource (TIMER) database. GSE14520 dataset and 80 HCC patients were used to validated the expression and survival value of COMMD3. Human HCC cell lines were also used for validating the function of COMMD3. RESULTS The expression of all COMMD family members showed higher expression in HCC tissues than that in normal tissues, and is associated with clinical cancer stage and pathological tumor grade. In HCC patients, the transcriptional levels of COMMD1/4 are positively correlated with overall survival (OS), while those of COMMD2/3/7/8/9 are negatively correlated with OS. Multivariate analysis indicated that a high level of COMMD3 mRNA is an independent prognostic factor for shorter OS in HCC patients. However, the subset of patients with grade 3 HCC, K-M survival curves revealed that high COMMD3/5/7/8/9 expression and low COMMD4/10 expression were associated with shorter OS. In addition, the expression of COMMD2/3/10 was associated with tumor-induced immune response activation and immune infiltration in HCC. The expression of COMMD3 from GSE14520 dataset and 80 patients are both higher in tumor than that in normal tissue, and a higher level of COMMD3 mRNA is associated with shorter OS. Knockdown of COMMD3 inhibits human HCC cell lines proliferation in vitro. CONCLUSIONS Our study indicates that COMMD3 is an independent prognostic biomarker for the survival of HCC patients. COMMD3 supports the proliferation of HCC cells and contributes to the poor OS in HCC patients.
Collapse
Affiliation(s)
- Xiaobo Wang
- Organ Transplant Center, the First Affiliated Hospital, Sun Yat-Sen University, NO.58 Zhongshan Road, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, 510080, China.,Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, 510080, China
| | - Shujiao He
- Organ Transplant Center, the First Affiliated Hospital, Sun Yat-Sen University, NO.58 Zhongshan Road, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, 510080, China.,Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, 510080, China
| | - Xin Zheng
- Department of Orthopaedics, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shanzhou Huang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Honghui Chen
- Organ Transplant Center, the First Affiliated Hospital, Sun Yat-Sen University, NO.58 Zhongshan Road, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, 510080, China.,Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, 510080, China
| | - Huadi Chen
- Organ Transplant Center, the First Affiliated Hospital, Sun Yat-Sen University, NO.58 Zhongshan Road, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, 510080, China.,Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, 510080, China
| | - Weixin Luo
- Organ Transplant Center, the First Affiliated Hospital, Sun Yat-Sen University, NO.58 Zhongshan Road, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, 510080, China.,Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, 510080, China
| | - Zhiyong Guo
- Organ Transplant Center, the First Affiliated Hospital, Sun Yat-Sen University, NO.58 Zhongshan Road, Guangzhou, 510080, China. .,Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, 510080, China. .,Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, 510080, China.
| | - Xiaoshun He
- Organ Transplant Center, the First Affiliated Hospital, Sun Yat-Sen University, NO.58 Zhongshan Road, Guangzhou, 510080, China. .,Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, 510080, China. .,Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, 510080, China.
| | - Qiang Zhao
- Organ Transplant Center, the First Affiliated Hospital, Sun Yat-Sen University, NO.58 Zhongshan Road, Guangzhou, 510080, China. .,Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, 510080, China. .,Guangdong Provincial International Cooperation Base of Science and Technology, Guangzhou, 510080, China.
| |
Collapse
|
25
|
Li R, Xu CQ, Shen JX, Ren QY, Chen DL, Lin MJ, Huang RN, Li CH, Zhong RT, Luo ZH, Ji XY, Wu J. 4-Methoxydalbergione is a potent inhibitor of human astroglioma U87 cells in vitro and in vivo. Acta Pharmacol Sin 2021; 42:1507-1515. [PMID: 33311599 PMCID: PMC8379167 DOI: 10.1038/s41401-020-00560-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/19/2020] [Indexed: 02/05/2023]
Abstract
Astroglioma is the most common primary tumor in the central nervous system without effective treatment strategies. Temozolomide (TMZ) is a chemotherapeutic drug to treat astroglioma but exhibits low potency and has side effects. Therefore, there is an urgent need to develop new compounds to treat astroglioma. Dalbergia sissoo Roxb was the source of Dalbergia odorifera in traditional Chinese medicine (TCM) and has been clinically used as an anti-tumor medicine. 4-Methoxydalbergione (4MOD) is purified from Dalbergia sissoo Roxb., and shows an inhibitory effect on osteosarcoma, but its effects on astroglioma have not been reported. Here, we evaluate its anti-astroglioma effects on both in vitro and in vivo models. In cultured astroglioma U87 cells, 4MOD inhibited cell proliferation and induced cell apoptosis in a time- and concentration-dependent manner. Compared with TMZ, 4MOD exhibited a tenfold greater potency of anti-astroglioma effects. 4MOD effectively stalled the cell cycle in G2 phase. Transcriptome sequencing (RNA-seq) showed that 4MOD upregulated 158 genes and downregulated 204 genes that are mainly enriched in cell membrane, cell division, cell cycle, p53, TNF, and MAPK signaling pathways, which may underlie its anti-tumor mechanisms. In a nude mouse xenograft model transplanted with U87 cells, 10 mg/kg 4MOD slowed down tumor growth rate, while at 30 mg/kg dose, it reduced tumor size. Collectively, this study demonstrates that 4MOD is a potent native compound that remarkably inhibits U87 astroglioma growth in both in vitro and in vivo models.
Collapse
Affiliation(s)
- Ran Li
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Chang-Qiong Xu
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Jian-Xin Shen
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Qiu-Yun Ren
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Di-Ling Chen
- Guangdong Institute of Microbiology, Guangzhou, 510070, China
| | - Mian-Jie Lin
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Rong-Ni Huang
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Chun-Hui Li
- Yueyang Hospital of Traditional Chinese Medicine, Yueyang, 414000, China
| | - Ru-Ting Zhong
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Zhi-Hua Luo
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Xiao-Yu Ji
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, 515041, China.
| | - Jie Wu
- Brain Function and Disease Laboratory, Shantou University Medical College, Shantou, 515041, China.
| |
Collapse
|
26
|
Thoms HC, Stark LA. The NF-κB Nucleolar Stress Response Pathway. Biomedicines 2021; 9:biomedicines9091082. [PMID: 34572268 PMCID: PMC8471347 DOI: 10.3390/biomedicines9091082] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/09/2021] [Accepted: 08/20/2021] [Indexed: 12/20/2022] Open
Abstract
The nuclear organelle, the nucleolus, plays a critical role in stress response and the regulation of cellular homeostasis. P53 as a downstream effector of nucleolar stress is well defined. However, new data suggests that NF-κB also acts downstream of nucleolar stress to regulate cell growth and death. In this review, we will provide insight into the NF-κB nucleolar stress response pathway. We will discuss apoptosis mediated by nucleolar sequestration of RelA and new data demonstrating a role for p62 (sequestosome (SQSTM1)) in this process. We will also discuss activation of NF-κB signalling by degradation of the RNA polymerase I (PolI) complex component, transcription initiation factor-IA (TIF-IA (RRN3)), and contexts where TIF-IA-NF-κB signalling may be important. Finally, we will discuss how this pathway is targeted by aspirin to mediate apoptosis of colon cancer cells.
Collapse
|
27
|
Li C, Peng H, Kang YJ. Cardiomyocyte-Specific COMMD1 Deletion Suppresses Ischemia-Induced Myocardial Apoptosis. Cardiovasc Toxicol 2021; 21:572-581. [PMID: 33900545 DOI: 10.1007/s12012-021-09650-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/07/2021] [Indexed: 02/05/2023]
Abstract
Copper metabolism MURR domain 1 (COMMD1) increases in ischemic myocardium along with suppressed contractility. Cardiomyocyte-specific deletion of COMMD1 preserved myocardial contractile function in response to the same ischemic insult. This study was undertaken to test the hypothesis that cardiomyocyte protection in COMMD1 myocardium is responsible for the functional preservation of the heart in response to ischemic insult. After ischemic insult, there were significantly more cardiomyocytes in the cardiomyocyte-specific COMMD1 deletion myocardium than that in WT controls. This preservation of cardiomyocytes was paralleled by a significant suppression of apoptosis in the COMMD1 deletion myocardium compared to controls. In searching for the mechanistic understanding of the anti-apoptotic effect of COMMD1 deletion, we found the anti-apoptotic Bcl-2 mRNA and protein expression were upregulated and the pro-apoptotic Bax mRNA and protein expression were downregulated. The critical transcription factor RelA, maintaining a high ratio between Bcl-2 and Bax for anti-apoptotic action, was suppressed by ischemia, but was rescued in the COMMD1 deletion myocardium. Because COMMD1 is critically involved in RelA ubiquitination and degradation, the data obtained here demonstrate that COMMD1 deletion leads to RelA preservation in ischemic myocardium, promoting the Bcl-2 anti-apoptotic pathway and suppressing the Bax pro-apoptotic pathway, and in combination, leading to protection of cardiomyocytes from ischemia-induced apoptosis.
Collapse
Affiliation(s)
- Chen Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hongxu Peng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Y James Kang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Memphis Institute of Regenerative Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
28
|
Abstract
Urate is the end-product of the purine metabolism in humans. The dominant source of urate is endogenous purines and the remainder comes through diet. Approximately two thirds of urate is eliminated via the kidney with the rest excreted in the feces. While the transporter BCRP, encoded by ABCG2, has been found to play a role in both the gut and kidney, SLC22A12 and SLC2A9 encoding URAT1 and GLUT9, respectively, are the two transporters best characterized. Only 8-12% of the filtered urate is excreted by the kidney. Renal elimination of urate depends substantially on specific transporters, including URAT1, GLUT9 and BCRP. Studies that have assessed the biologic effects of urate have produced highly variable results. Although there is a suggestion that urate may have anti-oxidant properties in some circumstances, the majority of evidence indicates that urate is pro-inflammatory. Hyperuricemia can result in the formation of monosodium urate (MSU) crystals that may be recognized as danger signals by the immune system. This immune response results in the activation of the NLRP3 inflammasome and ultimately in the production and release of interleukin-1β, and IL-18, that mediate both inflammation, pyroptotic cell death, and necroinflammation. It has also been demonstrated that soluble urate mediates effects on the kidney to induce hypertension and can induce long term epigenetic reprogramming in myeloid cells to induce "trained immunity." Together, these sequelae of urate are thought to mediate most of the physiological effects of hyperuricemia and gout, illustrating this biologically active molecule is more than just an "end-product" of purine metabolism.
Collapse
Affiliation(s)
- Robert T Keenan
- Division of Rheumatology, Duke University School of Medicine, Durham 27710, NC, USA.
| |
Collapse
|
29
|
Han F, Pang S, Sun Z, Cui Y, Yan B. Genetic Variants and Functional Analyses of the ATG16L1 Gene Promoter in Acute Myocardial Infarction. Front Genet 2021; 12:591954. [PMID: 34220924 PMCID: PMC8248370 DOI: 10.3389/fgene.2021.591954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 04/23/2021] [Indexed: 01/01/2023] Open
Abstract
Background Acute myocardial infarction (AMI), a common complex disease caused by an interaction between genetic and environmental factors, is a serious type of coronary artery disease and is also a leading cause of death worldwide. Autophagy-related 16-like 1 (ATG16L1) is a key regulatory factor of autophagy and plays an important role in induced autophagy. In the cardiovascular system, autophagy is essential to preserve the homeostasis and function of the heart and blood vessels. No studies have hitherto examined the association between AMI and ATG16L1 gene promoter. Methods We conducted a case-control study, using polymerase chain reaction and sequencing techniques, dual luciferase reporter assay, and electrophoretic mobility shift assay, to analyze genetic and functional variation in the ATG16L1 gene promoter between AMI and controls. A variety of statistical analyses were used to analyze the allele and genotype frequencies and the relationship between single-nucleotide polymorphisms (SNPs) and AMI. Results In all, 10 SNPs and two DNA-sequence variants (DSVs) were identified in 688 subjects, and three ATG16L1 gene promoter mutations [g.233250693 T > C (rs185213911), g.233250946 G > A (rs568956599), g.233251133 C > G (rs1301744254)] that were identified in AMI patients significantly altered the transcriptional activity of ATG16L1 gene promoter in HEH2, HEK-293, and H9c2 cells (P < 0.05). Further electrophoretic mobility shift assays indicated that the SNPs affected the binding of transcription factors (P < 0.01). Conclusion ATG16L1 gene promoter mutations in AMI patients may affect the binding of transcription factors and change the transcriptional activity of the ATG16L1 gene, changing the level of autophagy and contributing to the occurrence and development of AMI as rare and low-frequency risk factors.
Collapse
Affiliation(s)
- Falan Han
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Zhaoqing Sun
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yinghua Cui
- Division of Cardiology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,The Center for Molecular Genetics of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| |
Collapse
|
30
|
Wang W, Wang C, Chen W, Ding S. Advances in immunological research of amphioxus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 118:103992. [PMID: 33387559 DOI: 10.1016/j.dci.2020.103992] [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: 10/12/2020] [Revised: 12/17/2020] [Accepted: 12/26/2020] [Indexed: 06/12/2023]
Abstract
Amphioxus, one of the most closely related invertebrates to vertebrates, is an important animal model for studying the origin and evolution of vertebrate immunity, especially the transition from innate immunity to adaptive immunity. The current research progresses of amphioxus in the field of immune organs, immune cells, complement system, cytokines, nuclear factor kappa B, immune-related lectins and enzymes are summarized, and some issues that remain to be understood or are in need of further clarification are highlighted. We hope to provide references for more in-depth study of the amphioxus immune system and lay a solid foundation for the construction of three-dimensional immune network in amphioxus from ontogeny to phylogeny.
Collapse
Affiliation(s)
- Wenjun Wang
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China
| | - Changliu Wang
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China.
| | - Wei Chen
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China; Yantai Productivity Promotion Center, Yantai, 264003, People's Republic of China
| | - Shuo Ding
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China
| |
Collapse
|
31
|
Weiskirchen R, Penning LC. COMMD1, a multi-potent intracellular protein involved in copper homeostasis, protein trafficking, inflammation, and cancer. J Trace Elem Med Biol 2021; 65:126712. [PMID: 33482423 DOI: 10.1016/j.jtemb.2021.126712] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/10/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022]
Abstract
Copper is a trace element indispensable for life, but at the same time it is implicated in reactive oxygen species formation. Several inherited copper storage diseases are described of which Wilson disease (copper overload, mutations in ATP7B gene) and Menkes disease (copper deficiency, mutations in ATP7A gene) are the most prominent ones. After the discovery in 2002 of a novel gene product (i.e. COMMD1) involved in hepatic copper handling in Bedlington terriers, studies on the mechanism of action of COMMD1 revealed numerous non-copper related functions. Effects on hepatic copper handling are likely mediated via interactions with ATP7B. In addition, COMMD1 has many more interacting partners which guide their routing to either the plasma membrane or, often in an ubiquitination-dependent fashion, trigger their proteolysis via the S26 proteasome. By stimulating NF-κB ubiquitination, COMMD1 dampens an inflammatory reaction. Finally, targeting COMMD1 function can be a novel approach in the treatment of tumors.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital Aachen, Aachen, Germany
| | - Louis C Penning
- Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Department of Clinical Sciences of Companion Animals, 3584 CM, Utrecht, the Netherlands.
| |
Collapse
|
32
|
Corbee RJ, Penning LC. COMMD1 Exemplifies the Power of Inbred Dogs to Dissect Genetic Causes of Rare Copper-Related Disorders. Animals (Basel) 2021; 11:ani11030601. [PMID: 33668783 PMCID: PMC7996361 DOI: 10.3390/ani11030601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Wilson's Disease is a rare autosomal recessive disorder in humans, often presenting with hepatic copper overload. Finding the genetic cause of a rare disease, especially if it is related to food constituents like the trace element copper, is a Herculean task. This review describes examples of how the unique population structure of in-bred dog strains led to the discovery of a novel gene and two modifier genes involved in inherited copper toxicosis. COMMD1, after the discovery in 2002, was shown to be a highly promiscuous protein involved in copper transport, protein trafficking/degradation, regulation of virus replication, and inflammation. Mutations in the ATP7A and ATP7B proteins in Labrador retrievers and Dobermann dogs resulted in a wide variation in hepatic copper levels in these breeds. To our knowledge, numerous dog breeds with inherited copper toxicosis of unknown genetic origin exist. Therefore, the possibility that men's best friend will provide new leads in rare copper storage diseases seems realistic.
Collapse
|
33
|
The Anticancer Peptide CIGB-552 Exerts Anti-Inflammatory and Anti-Angiogenic Effects through COMMD1. Molecules 2020; 26:molecules26010152. [PMID: 33396282 PMCID: PMC7795859 DOI: 10.3390/molecules26010152] [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] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/24/2022] Open
Abstract
CIGB-552 is a synthetic anti-tumor peptide capable of reducing tumor size and increasing the lifespan of tumor-bearing mice. Part of its anti-cancer effects consists of inducing apoptosis, modulating NF-kB signaling pathway, and the angiogenesis process. Although one of its major mediators, the COMMD1 protein, has been identified, the mechanism by which CIGB-552 exerts such effects remains elusive. In the present study, we show the role of COMMD1 in CIGB-552 mechanism of action by generating the COMMD1 knock-out from the human lung cancer cell line NCI-H460. A microarray was performed to analyze both wild-type and KO cell lines with regard to CIGB-552 treatment. Additionally, different signaling pathways were studied in both cell lines to validate the results. Furthermore, the interaction between CIGB-552 and COMMD1 was analyzed by confocal microscopy. By signaling pathway analysis we found that genes involved in cell proliferation and apoptosis, oncogenic transformation, angiogenesis and inflammatory response are potentially regulated by the treatment with CIGB-552. We then demonstrated that CIGB-552 is capable of modulating NF-kB in both 2D and 3D cell culture models. Finally, we show that the ability of CIGB-552 to negatively modulate NF-kB and HIF-1 pathways is impaired in the COMMD1 knock-out NCI-H460 cell line, confirming that COMMD1 is essential for the peptide mechanism of action.
Collapse
|
34
|
Oh GS, Yoon J, Kim G, Kim GH, Kim DS, Choi B, Chang EJ, Lee ES, Kim SW. Regulation of adipocyte differentiation by clusterin-mediated Krüppel-like factor 5 stabilization. FASEB J 2020; 34:16276-16290. [PMID: 33078455 DOI: 10.1096/fj.202000551rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 09/25/2020] [Accepted: 10/01/2020] [Indexed: 12/26/2022]
Abstract
Clusterin (CLU) is a heterodimeric glycoprotein involved in a range of biological processes. We investigated the function of CLU as a novel regulator of adipogenesis. CLU expression increased during 3T3-L1 preadipocyte differentiation. CLU overexpression promoted adipogenic differentiation of preadipocytes and increased the mRNA levels of adipogenic markers including peroxisome proliferator-activated receptor γ (Pparg) and CCAAT enhancer-binding protein α (Cebpa). Conversely, knockdown of CLU attenuated adipogenesis and reduced transcript levels of Pparg and Cebpa. However, the promoter activities of both the Pparg and the Cebpa gene were not affected by alteration of CLU expression on its own. Additionally, the protein level of Krüppel-like factor 5 (KLF5), an upstream transcription factor of Pparg and Cebpa involved in adipogenic differentiation, was upregulated by CLU overexpression, although the mRNA level of Klf5 was not altered by changes in the expression level of CLU. Cycloheximide chase assay showed that the increased level of KLF5 by CLU overexpression was due to decreased degradation of KLF5 protein. Interestingly, CLU increased the stability of KLF5 by decreasing KLF5 ubiquitination. CLU inhibited the interaction between KLF5 and F-box/WD repeat-containing protein 7, which is an E3 ubiquitin ligase that targets KLF5. The adipogenic role of CLU was also addressed in mesenchymal stem cells (MSCs) and Clu-/- mouse embryonic fibroblasts (MEFs). Furthermore, CLU enhanced KLF5-mediated transcriptional activation of both the Cebpa and the Pparg promoter. Taken together, these results suggest that CLU is a novel regulator of adipocyte differentiation by modulating the protein stability of the adipogenic transcription factor KLF5.
Collapse
Affiliation(s)
- Gyun-Sik Oh
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Bio-Medical Institute of Technology, University of Ulsan, Seoul, Republic of Korea
| | - Jin Yoon
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Bio-Medical Institute of Technology, University of Ulsan, Seoul, Republic of Korea
| | - Gukhan Kim
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Geun Hyang Kim
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong Seop Kim
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Bio-Medical Institute of Technology, University of Ulsan, Seoul, Republic of Korea.,Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bongkun Choi
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun-Ju Chang
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun-Sook Lee
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Bio-Medical Institute of Technology, University of Ulsan, Seoul, Republic of Korea
| | - Seung-Whan Kim
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Bio-Medical Institute of Technology, University of Ulsan, Seoul, Republic of Korea.,Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
35
|
Chen W, Wang J, Hua Z, Zhang Y. Du Huo Ji Sheng Tang relieves knee osteoarthritis via suppressing NLRP3/NF-κB inflammatory signals in rats. EUR J INFLAMM 2020. [DOI: 10.1177/2058739220942627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Knee osteoarthritis (KOA) is a common chronic disease in the elderly and leads to a high rate of disability. Du Huo Ji Sheng Tang (DHJST), a Chinese traditional medicinal formula, is a classic prescription for the treatment of KOA. Here, we investigated whether DHJST could inhibit inflammation and treat KOA through suppressing NLRP3/nuclear factor (NF)-κB inflammatory signals in rats. The serum levels of interleukin (IL)-1β, IL-6, IL-10, tumor necrosis factor (TNF)-α, NLRP3, ASC, Caspase-1, p-NF-κB-P65, and p-IκBa were detected in healthy adults and patients with KOA before and after treatment. Sprague Dawley (SD) rats were divided into normal group, model group, diclofenac sodium group (5 mg/kg), DHJST high-dose group (1 g/kg), and DHJST low-dose group (0.5 g/kg). The right hind knee joint of the rats, except normal group, was injected with 4% papain (0.25 mL/kg) once every 7 days for three times. All rats were treated for 3 weeks. The swelling volume of right hind paw; five classification of inflammatory cells in synovial fluid; pathological changes of the knee-joint synovial membrane and cartilage; levels of IL-1β, IL-6, and TNF-α in serum and knee-joint synovial fluid; and the expressions of NLRP3/NF-κB inflammatory signals in the knee-joint synovial membrane were detected. The serum levels of IL-1β, IL-6, IL-10, TNF-α, NLRP3, ASC, Caspase-1, p-NF-κB-P65, and p-IκBa in KOA patients treated with DHJST were significantly decreased. The KOA rats treated with DHJST showed significant decreases in swelling volume of right hind paws; the percentage of leukocyte, lymphocyte, neutrophil, and eosinophils in synovial fluid; the levels of IL-1β, IL-6, and TNF-α in serum and knee-joint synovial fluid; and the expressions of NLRP3 ASC, Caspase-1, IL-1β, p-NF-κB-P65, and p-IκBa in the knee-joint synovial membrane, and showed an alleviation in pathological changes of the knee-joint synovial membrane and cartilage. Our data provide the first evidence that DHJST relieves KOA via suppressing NLRP3/NF-κB inflammatory signals in rats
Collapse
Affiliation(s)
- Wenjin Chen
- Department of Orthopedics, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Jianwei Wang
- Department of Orthopedics, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Zhen Hua
- Department of Orthopedics, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Yafeng Zhang
- Department of Orthopedics, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
36
|
Yang M, Huang W, Sun Y, Liang H, Chen M, Wu X, Wang X, Zhang L, Cheng X, Fan Y, Pan H, Chen L, Guan J. Prognosis and modulation mechanisms of COMMD6 in human tumours based on expression profiling and comprehensive bioinformatics analysis. Br J Cancer 2019; 121:699-709. [PMID: 31523056 PMCID: PMC6889128 DOI: 10.1038/s41416-019-0571-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 08/05/2019] [Accepted: 08/21/2019] [Indexed: 12/24/2022] Open
Abstract
Background The Copper Metabolism MURR1 (COMM) domain family has been reported to play important roles in tumorigenesis. As a prototype for the COMMD family, the expression pattern and biological function of COMMD6 in human tumours remain unknown. Methods COMMD6 expression in BALB/c mice and human tissues was examined using real-time PCR and immunohistochemistry. Kaplan–Meier analysis was applied to evaluate the prognosis of COMMD6 in tumours. Competing endogenous RNA (ceRNA) and transcriptional regulation network were constructed based on differentially expressed mRNAs, microRNAs and long non-coding RNAs from the cancer genome atlas database. GO and KEGG enrichment analysis were used to explore the bioinformatics implication. Results COMMD6 expression was widely observed in BALB/c mice and human tissues, which predicted prognosis of cancer patients. Furthermore, we shed light on the underlying tumour promoting role and mechanism of COMMD6 by constructing a TEX41-miR-340-COMMD6 ceRNA network in head and neck squamous cell carcinoma and miR-218-CDX1-COMMD6 transcriptional network in cholangiocarcinoma. In addition, COMMD6 may modulate the ubiquitination and degradation of NF-κB subunits and regulate ribonucleoprotein and spliceosome complex biogenesis in tumours. Conclusions This study may help to elucidate the functions and mechanisms of COMMD6 in human tumours, providing a potential biomarker for tumour prevention and therapy.
Collapse
Affiliation(s)
- Mi Yang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China
| | - Weiqiang Huang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China
| | - Yaling Sun
- Department of Radiation Oncology, The Third Affiliated Hospital, Sun Yat-sen University, Guangdong, Guangzhou, China
| | - Huazhen Liang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China
| | - Min Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China
| | - Xixi Wu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China
| | - Xiaoqing Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China
| | - Longshan Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China
| | - Xiaoya Cheng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China
| | - Yao Fan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China
| | - Hua Pan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China
| | - Longhua Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China.
| | - Jian Guan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangdong, Guangzhou, China.
| |
Collapse
|
37
|
De Luca A, Barile A, Arciello M, Rossi L. Copper homeostasis as target of both consolidated and innovative strategies of anti-tumor therapy. J Trace Elem Med Biol 2019; 55:204-213. [PMID: 31345360 DOI: 10.1016/j.jtemb.2019.06.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/28/2019] [Accepted: 06/14/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Copper was reported to be involved in the onset and progression of cancer. Proteins in charge of copper uptake and distribution, as well as cuproenzymes, are altered in cancer. More recently, proteins involved in signaling cascades, regulating cell proliferation, and anti-apoptotic protein factors were found to interact with copper. Therefore, therapeutic strategies using copper complexing molecules have been proposed for cancer therapy and used in clinical trials. OBJECTIVES This review will focus on novel findings about the involvement of copper and cupro-proteins in cancer dissemination process, epithelium to mesenchymal transition and vascularization. Particularly, implication of well-established (e.g. lysil oxidase) or newly identified copper-binding proteins (e.g. MEMO1), as well as their interplay, will be discussed. Moreover, we will describe recently synthesized copper complexes, including plant-derived ones, and their efficacy in contrasting cancer development. CONCLUSIONS The research on the involvement of copper in cancer is still an open field. Further investigation is required to unveil the mechanisms involved in copper delivery to the novel copper-binding proteins, which may identify other possible gene and protein targets for cancer therapy.
Collapse
Affiliation(s)
| | - Anna Barile
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| | - Mario Arciello
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| | - Luisa Rossi
- Department of Biology, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
38
|
Umbreen S, Banday MM, Jamroze A, Mansini AP, Ganaie AA, Ferrari MG, Maqbool R, Beigh FH, Murugan P, Morrissey C, Corey E, Konety BR, Saleem M. COMMD3:BMI1 Fusion and COMMD3 Protein Regulate C-MYC Transcription: Novel Therapeutic Target for Metastatic Prostate Cancer. Mol Cancer Ther 2019; 18:2111-2123. [PMID: 31467179 DOI: 10.1158/1535-7163.mct-19-0150] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/24/2019] [Accepted: 08/20/2019] [Indexed: 11/16/2022]
Abstract
Gene rearrangement is reported to be associated to the aggressive phenotype and poor prognosis in prostate cancer. We identified a gene fusion between a transcription repressor (BMI1) and transcriptional factor (COMMD3) in human prostate cancer. We show that COMMD3:BMI1 fusion expression is significantly increased in prostate cancer disease in an order: normal tissue < primary < metastatic tumors (Mets). Although elevated TMPRSS-ERG/ETV fusion is reported in prostate cancer, we identified a subtype of Mets exhibiting low TMPRSS:ETV and high COMMD3:BMI1 We delineated the mechanism and function of COMMD3 and COMMD3:BMI1 in prostate cancer. We show that COMMD3 level is elevated in prostate cancer cell models, PDX models (adenocarcinoma, NECaP), and Mets. The analysis of TCGA/NIH/GEO clinical data showed a positive correlation between increased COMMD3 expression to the disease recurrence and poor survival in prostate cancer. We show that COMMD3 drives proliferation of normal cells and promotes migration/invasiveness of neoplastic cells. We show that COMMD3:BMI1 and COMMD3 regulate C-MYC transcription and C-MYC downstream pathway. The ChIP analysis showed that COMMD3 protein is recruited at the promoter of C-MYC gene. On the basis of these data, we investigated the relevance of COMMD3:BMI1 and COMMD3 as therapeutic targets using in vitro and xenograft mouse models. We show that siRNA-mediated targeting of COMMD3:BMI1 and COMMD3 significantly decreases (i) C-MYC expression in BRD/BET inhibitor-resistant cells, (ii) proliferation/invasion in vitro, and (iii) growth of prostate cancer cell tumors in mice. The IHC analysis of tumors confirmed the targeting of COMMD3-regulated molecular pathway under in vivo conditions. We conclude that COMMD3:BMI1 and COMMD3 are potential progression biomarkers and therapeutic targets of metastatic prostate cancer.
Collapse
Affiliation(s)
- Syed Umbreen
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Queens University, Belfast, Northern Ireland
| | - Mudassir Meraj Banday
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Anmbreen Jamroze
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Hormel Institute, Austin, Minnesota
| | - Adrian P Mansini
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Arsheed A Ganaie
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Marina G Ferrari
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Raihana Maqbool
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Firdous H Beigh
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | | | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, Washington
| | - Badrinath R Konety
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Mohammad Saleem
- Department of Urology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
39
|
Mallam AL, Marcotte EM. Systems-wide Studies Uncover Commander, a Multiprotein Complex Essential to Human Development. Cell Syst 2019; 4:483-494. [PMID: 28544880 DOI: 10.1016/j.cels.2017.04.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/25/2017] [Accepted: 03/23/2017] [Indexed: 11/27/2022]
Abstract
Recent mass spectrometry maps of the human interactome independently support the existence of a large multiprotein complex, dubbed "Commander." Broadly conserved across animals and ubiquitously expressed in nearly every human cell type examined thus far, Commander likely plays a fundamental cellular function, akin to other ubiquitous machines involved in expression, proteostasis, and trafficking. Experiments on individual subunits support roles in endosomal protein sorting, including the trafficking of Notch proteins, copper transporters, and lipoprotein receptors. Commander is critical for vertebrate embryogenesis, and defects in the complex and its interaction partners disrupt craniofacial, brain, and heart development. Here, we review the synergy between large-scale proteomic efforts and focused studies in the discovery of Commander, describe its composition, structure, and function, and discuss how it illustrates the power of systems biology. Based on 3D modeling and biochemical data, we draw strong parallels between Commander and the retromer cargo-recognition complex, laying a foundation for future research into Commander's role in human developmental disorders.
Collapse
Affiliation(s)
- Anna L Mallam
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA.
| | - Edward M Marcotte
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
40
|
Jiang Z, Chen W, Zhou J, Peng Q, Zheng H, Yuan Y, Cui H, Zhao W, Sun X, Zhou Z, Liu X. Identification of COMMD1 as a novel lamin A binding partner. Mol Med Rep 2019; 20:1790-1796. [PMID: 31257505 PMCID: PMC6625409 DOI: 10.3892/mmr.2019.10419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 05/20/2019] [Indexed: 01/28/2023] Open
Abstract
Lamin A, which is encoded by the LMNA gene, regulates gene expression and genome stability through interactions with a variety of proteins. Mutations in LMNA lead to a diverse set of inherited human diseases, collectively referred to as laminopathies. To gain insight into the protein interactions of lamin A, a yeast two-hybrid screen was conducted using the carboxy-terminus of lamin A. The screen identified copper metabolism MURR1 domain-containing 1 (COMMD1) as a novel lamin A binding partner. Colocalization experiments using fluorescent confocal microscopy revealed that COMMD1 colocalized with lamin A in 293 cells. Furthermore, the COMMD1-lamin A protein interaction was also demonstrated in co-immunoprecipitation experiments. Collectively, the present study demonstrated a physical interaction between COMMD1 and lamin A, which may aid to elucidate the mechanisms of lamin A in the aging process.
Collapse
Affiliation(s)
- Zhiwen Jiang
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Weichun Chen
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Jing Zhou
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Qi Peng
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Huiling Zheng
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Yuan Yuan
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Hongjing Cui
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Wei Zhao
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Xuerong Sun
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Zhongjun Zhou
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Xinguang Liu
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
41
|
Chen K, Healy MD, Collins BM. Towards a molecular understanding of endosomal trafficking by Retromer and Retriever. Traffic 2019; 20:465-478. [DOI: 10.1111/tra.12649] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Kai‐En Chen
- Institute for Molecular Bioscience University of Queensland St. Lucia Queensland Australia
| | - Michael D. Healy
- Institute for Molecular Bioscience University of Queensland St. Lucia Queensland Australia
| | - Brett M. Collins
- Institute for Molecular Bioscience University of Queensland St. Lucia Queensland Australia
| |
Collapse
|
42
|
Wang J, Fan SM, Zhang J. Epigallocatechin-3-gallate ameliorates lipopolysaccharide-induced acute lung injury by suppression of TLR4/NF-κB signaling activation. ACTA ACUST UNITED AC 2019; 52:e8092. [PMID: 31241712 PMCID: PMC6596362 DOI: 10.1590/1414-431x20198092] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 05/02/2019] [Indexed: 12/27/2022]
Abstract
Acute lung injury (ALI) is a serious clinical syndrome with a high rate of mortality. The activation of inflammation is well-recognized as a vital factor in the pathogenesis of lipopolysaccharide (LPS)-induced ALI. Therefore, suppression of the inflammatory response could be an ideal strategy to prevent ALI. Epigallocatechin-3-gallate (EGCG), mainly from green tea, has been shown to have an anti-inflammatory effect. The aim of the study was to explore whether EGCG alleviates inflammation in sepsis-related ALI. Male BALB/C mice were treated with EGCG (10 mg/kg) intraperitoneally (ip) 1 h before LPS injection (10 mg/kg, ip). The results showed that EGCG attenuated LPS-induced ALI as it decreased the changes in blood gases and reduced the histological lesions, wet-to-dry weight ratios, and myeloperoxidase (MPO) activity. In addition, EGCG significantly decreased the expression of pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 in the lung, serum, and bronchoalveolar lavage fluid, and alleviated the expression of TLR-4, MyD88, TRIF, and p-p65 in the lung tissue. In addition, it increased the expression of IκB-α and had no influence on the expression of p65. Collectively, these results demonstrated the protective effects of EGCG against LPS-induced ALI in mice through its anti-inflammatory effect that may be attributed to the suppression of the activation of TLR 4-dependent NF-κB signaling pathways.
Collapse
Affiliation(s)
- Jia Wang
- General Practice Center, University of Electronic Science and Technology, Sichuan Academy of Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Shi Ming Fan
- Department of Respiratory Medicine, Changning Hospital of Traditional Chinese Medicine, Yibin, China
| | - Jiong Zhang
- Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| |
Collapse
|
43
|
Ji YP, Shi TY, Zhang YY, Lin D, Linghu KG, Xu YN, Tao L, Lu Q, Shen XC. Essential oil from Fructus Alpinia zerumbet (fruit of Alpinia zerumbet (Pers.) Burtt.et Smith) protected against aortic endothelial cell injury and inflammation in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2019; 237:149-158. [PMID: 30880260 DOI: 10.1016/j.jep.2019.03.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 03/05/2019] [Accepted: 03/05/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fructus Alpinia zerumbet (FAZ), a dry and ripe fruit of Alpinia zerumbet (Pers.) Burtt. et Smith, is widely used as a spice to treat cardiovascular diseases in clinic as a miao folk medicine in Guizhou Province of China. Essential oil extracted from FAZ (EOFAZ) is the key bioactive ingredients. AIM OF THE STUDY This study aimed to examine the effects and mechanisms of EOFAZ on lipopolysaccharide (LPS)-induced endothelial cell injury, inflammation and apoptosis in vitro and in vivo. MATERIALS AND METHODS For the in vitro study, LPS-treated human aortic endothelial cells were used to perform PCR, western blot analysis and immunofluorescence. For the in vivo study, male mouse were divided into four groups, vehicle control group and LPS group received 0.5% Tween-80 in saline; and two EOFAZ groups receive different dose of EOFAZ (90 mg kg -1·day-1, 180 mg kg -1·day-1) respectively. Each group was fed for 7 days by intragastrical administration at daily base. Then, except vehicle control group received saline, mice in other three groups were administered with LPS (1 mg kg -1, dissolved in saline) by intraperitoneal injection. 24 h later, Aorta tissue was collected and frozen immediately in liquid N2, stored at -80 °C for western blot analysis. RESULTS We found that EOFAZ completely prevented LPS-induced HAEC activation and inflammation in vitro and in vivo, as assessed by expression of endothelial adhesion molecules, ICAM-1 and VCAM-1. Similarly, EOFAZ significantly blunted LPS-induced endothelial injury, as tested by MTT assay, LDH release and caspase-3 activation. We further demonstrated that TLR4-dependent NF-κB signaling may be involved in the process. CONCLUSION EOFAZ protected against LPS-induced endothelial cell injury and inflammation likely via inhibition of TLR4-dependent NF-κB signaling.
Collapse
Affiliation(s)
- Yun-Peng Ji
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China; Department of Pharmacy, Guizhou Provincial People's Hospital, Guiyang City, Guizhou Province, China
| | - Ting-Yu Shi
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China; Department of Neonatal Disease Screening, Shenyang Maternity and Child Health Hospital, Shenyang City, Liaoning Province, China
| | - Yan-Yan Zhang
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Dan Lin
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Ke-Gang Linghu
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Yi-Ni Xu
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Ling Tao
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center/ Alpert Medical School of Brown University, Providence, RI, USA.
| | - Xiang-Chun Shen
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China.
| |
Collapse
|
44
|
CRISP-R/Cas9 Mediated Deletion of Copper Transport Genes CTR1 and DMT1 in NSCLC Cell Line H1299. Biological and Pharmacological Consequences. Cells 2019; 8:cells8040322. [PMID: 30959888 PMCID: PMC6523758 DOI: 10.3390/cells8040322] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 12/27/2022] Open
Abstract
Copper, the highly toxic micronutrient, plays two essential roles: it is a catalytic and structural cofactor for Cu-dependent enzymes, and it acts as a secondary messenger. In the cells, copper is imported by CTR1 (high-affinity copper transporter 1), a transmembrane high-affinity copper importer, and DMT1 (divalent metal transporter). In cytosol, enzyme-specific chaperones receive copper from CTR1 C-terminus and deliver it to their apoenzymes. DMT1 cannot be a donor of catalytic copper because it does not have a cytosol domain which is required for copper transfer to the Cu-chaperons that assist the formation of cuproenzymes. Here, we assume that DMT1 can mediate copper way required for a regulatory copper pool. To verify this hypothesis, we used CRISPR/Cas9 to generate H1299 cell line with CTR1 or DMT1 single knockout (KO) and CTR1/DMT1 double knockout (DKO). To confirm KOs of the genes qRT-PCR were used. Two independent clones for each gene were selected for further studies. In CTR1 KO cells, expression of the DMT1 gene was significantly increased and vice versa. In subcellular compartments of the derived cells, copper concentration dropped, however, in nuclei basal level of copper did not change dramatically. CTR1 KO cells, but not DMT1 KO, demonstrated reduced sensitivity to cisplatin and silver ions, the agents that enter the cell through CTR1. Using single CTR1 and DMT1 KO, we were able to show that both, CTR1 and DMT1, provided the formation of vital intracellular cuproenzymes (SOD1, COX), but not secretory ceruloplasmin. The loss of CTR1 resulted in a decrease in the level of COMMD1, XIAP, and NF-κB. Differently, the DMT1 deficiency induced increase of the COMMD1, HIF1α, and XIAP levels. The possibility of using CTR1 KO and DMT1 KO cells to study homeodynamics of catalytic and signaling copper selectively is discussed.
Collapse
|
45
|
Expression profile and bioinformatics analysis of COMMD10 in BALB/C mice and human. Cancer Gene Ther 2019; 27:216-225. [PMID: 30787448 DOI: 10.1038/s41417-019-0087-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/22/2019] [Accepted: 01/26/2019] [Indexed: 11/08/2022]
Abstract
COMMD10, a member of COMMD protein, has been proved to target p65 NF-kappaB (nuclear factor-kappaB) subunit and reduce its nuclear translocation, thereby leading to the inactivation of NF-kappaB pathway and suppression of colorectal cancer invasion and metastasis. The aim of this study is to explore its expression pattern and tissue distribution in human normal tissues and other tumor tissues and to investigate the relevant mechanism. We firstly provided the expression profile and histological distribution of COMMD10 in various BALB/c mice tissues and identified the biological distribution of COMMD10 in different kinds of human normal and tumor tissues. We verified the expression profile of COMMD10 using TCGA database. The interacting genes of COMMD10 were predicted by using STRING using. Finally, we performed database, and the microRNAs targeting COMMD10 were predicted using miRDB, miRWalk, TargetScan and microRNA. GO and KEGG pathway analyses were performed to predict the biological function of COMMD10 and its interacting genes. mRNA expression of COMMD10 showed the highest level in the lung and spleen, and the lowest level in the heart and brain. Immunohistochemistry detection revealed that COMMD10 was expressed in different tissues with different degrees and was was located mainly in the cytoplasm. Subsequently, we showed that COMMD10 displayed various degrees of expression in different human normal tissues that mainly located in cytoplasm, while COMMD10 of liver cells resided in both nucleus and cytoplasm. All the tumor tissues except breast small cell carcinoma, breast phyllodes tumor, lung adenocarcinoma, thymoma, cervical cancer and bladder urothelial carcinoma showed that COMMD10 was positive staining in cytoplasm. Kaplan-Meier plotter indicated that renal clear cell carcinoma patients with increased expression level of COMMD10 exhibited longer survival. STRING database revealed that COMMD10 had 41 interacting genes, and data from 4 different databases indicated that hsa-miR-590-3p may be the potential regulator of COMMD10. GO analysis demonstrated that COMMD10 and its interacting genes were mainly enriched in Cullin-RING ubiquitin ligase complexes, binding and transport of copper ions, the transport and steady-state maintenance of copper ions, transcription, translation and transport of proteins, and negatively regulate the activity of NF-kappaB transcription factors. KEGG pathway showed that COMMD10 and its interacting genes were mainly involved in renal cell carcinoma, HIF-1 signaling pathways, ubiquitination-mediated proteolysis, endocytosis and mineral absorption. COMMD10 may play a tumor suppressive role in renal clear cell carcinoma through the miR-590-3p-COMMD10-Cul2-RBX1-NF-κB/HIF/NRF2 pathway and regulate the chemotherapy resistance of various tumor cells to cisplatin.
Collapse
|
46
|
Composition of the Survival Motor Neuron (SMN) Complex in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2019; 9:491-503. [PMID: 30563832 PMCID: PMC6385987 DOI: 10.1534/g3.118.200874] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Spinal Muscular Atrophy (SMA) is caused by homozygous mutations in the human survival motor neuron 1 (SMN1) gene. SMN protein has a well-characterized role in the biogenesis of small nuclear ribonucleoproteins (snRNPs), core components of the spliceosome. SMN is part of an oligomeric complex with core binding partners, collectively called Gemins. Biochemical and cell biological studies demonstrate that certain Gemins are required for proper snRNP assembly and transport. However, the precise functions of most Gemins are unknown. To gain a deeper understanding of the SMN complex in the context of metazoan evolution, we investigated its composition in Drosophila melanogaster Using transgenic flies that exclusively express Flag-tagged SMN from its native promoter, we previously found that Gemin2, Gemin3, Gemin5, and all nine classical Sm proteins, including Lsm10 and Lsm11, co-purify with SMN. Here, we show that CG2941 is also highly enriched in the pulldown. Reciprocal co-immunoprecipitation reveals that epitope-tagged CG2941 interacts with endogenous SMN in Schneider2 cells. Bioinformatic comparisons show that CG2941 shares sequence and structural similarity with metazoan Gemin4. Additional analysis shows that three other genes (CG14164, CG31950 and CG2371) are not orthologous to Gemins 6-7-8, respectively, as previously suggested. In D.melanogaster, CG2941 is located within an evolutionarily recent genomic triplication with two other nearly identical paralogous genes (CG32783 and CG32786). RNAi-mediated knockdown of CG2941 and its two close paralogs reveals that Gemin4 is essential for organismal viability.
Collapse
|
47
|
Papadaki M, Rinotas V, Violitzi F, Thireou T, Panayotou G, Samiotaki M, Douni E. New Insights for RANKL as a Proinflammatory Modulator in Modeled Inflammatory Arthritis. Front Immunol 2019; 10:97. [PMID: 30804932 PMCID: PMC6370657 DOI: 10.3389/fimmu.2019.00097] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/14/2019] [Indexed: 01/01/2023] Open
Abstract
Receptor activator of nuclear factor-κB ligand (RANKL), a member of the Tumor Necrosis Factor (TNF) superfamily, constitutes the master regulator of osteoclast formation and bone resorption, whereas its involvement in inflammatory diseases remains unclear. Here, we used the human TNF transgenic mouse model of erosive inflammatory arthritis to determine if the progression of inflammation is affected by either genetic inactivation or overexpression of RANKL in transgenic mouse models. TNF-mediated inflammatory arthritis was significantly attenuated in the absence of functional RANKL. Notably, TNF overexpression could not compensate for RANKL-mediated osteopetrosis, but promoted osteoclastogenesis between the pannus and bone interface, suggesting RANKL-independent mechanisms of osteoclastogenesis in inflamed joints. On the other hand, simultaneous overexpression of RANKL and TNF in double transgenic mice accelerated disease onset and led to severe arthritis characterized by significantly elevated clinical and histological scores as shown by aggressive pannus formation, extended bone resorption, and massive accumulation of inflammatory cells, mainly of myeloid origin. RANKL and TNF cooperated not only in local bone loss identified in the inflamed calcaneous bone, but also systemically in distal femurs as shown by microCT analysis. Proteomic analysis in inflamed ankles from double transgenic mice overexpressing human TNF and RANKL showed an abundance of proteins involved in osteoclastogenesis, pro-inflammatory processes, gene expression regulation, and cell proliferation, while proteins participating in basic metabolic processes were downregulated compared to TNF and RANKL single transgenic mice. Collectively, these results suggest that RANKL modulates modeled inflammatory arthritis not only as a mediator of osteoclastogenesis and bone resorption but also as a disease modifier affecting inflammation and immune activation.
Collapse
Affiliation(s)
- Maria Papadaki
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece.,Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Vagelis Rinotas
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece.,Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Foteini Violitzi
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece.,Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Trias Thireou
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - George Panayotou
- Division of Molecular Oncology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Martina Samiotaki
- Division of Molecular Oncology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| | - Eleni Douni
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece.,Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Athens, Greece
| |
Collapse
|
48
|
Jiang Z, Yuan Y, Zheng H, Cui H, Sun X, Zhao W, Liu X. COMMD1 regulates cell proliferation and cell cycle progression by modulating p21 Cip1 levels. Biosci Biotechnol Biochem 2019; 83:845-850. [PMID: 30667321 DOI: 10.1080/09168451.2019.1569497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Copper metabolism MURR1 domain-containing 1 (COMMD1) is a protein that participates in multiple cellular processes, including copper homeostasis and nuclear factor kappa B (NF-κB) and hypoxia-inducible factor 1α (HIF-1α) signaling. The COMMD1 upstream regulators X-linked inhibitor of apoptosis protein (XIAP) and p300 and downstream targets such as NF-κB and HIF-1α are involved in the regulation of cell proliferation and cell cycle progression. However, whether COMMD1 regulates cell proliferation and the cell cycle remains unclear. In the present study, we demonstrated that both overexpression and knockdown of COMMD1 affected the proliferation of HEK293 cells, and the cell cycle assay revealed that ectopic expression of COMMD1 arrested the cell cycle at the G1 phase. Furthermore, western blot analysis showed that COMMD1 affected p21 Cip1 levels. Taken together, these results suggest that COMMD1 regulates cell proliferation and cell cycle progression by modulating p21 Cip1 levels. Abbreviations COMMD1: Copper metabolism MURR1 domain containing 1; XIAP: X chromosome-linked inhibitor of apoptosis protein; FCS: Fetal calf serum; WCE: Whole cell extracts; RT-PCR: Reverse transcription-polymerase chain reaction; HEK293: Human embryonic kidney 293; ShRNA: Short hairpin RNA; NF-κB: Nuclear factor kappa-light-chain-enhancer of activated B cells; ARF: Alternate reading frame protein product of the CDKN2A locus.
Collapse
Affiliation(s)
- Zhiwen Jiang
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Yuan Yuan
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Huiling Zheng
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Hongjing Cui
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Xuerong Sun
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Wei Zhao
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Xinguang Liu
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China.,c Institute of Biochemistry and Molecular Biology , Guangdong Medical University , Dongguan , China
| |
Collapse
|
49
|
Zheng L, You N, Huang X, Gu H, Wu K, Mi N, Li J. COMMD7 Regulates NF-κB Signaling Pathway in Hepatocellular Carcinoma Stem-like Cells. MOLECULAR THERAPY-ONCOLYTICS 2018; 12:112-123. [PMID: 30719501 PMCID: PMC6350112 DOI: 10.1016/j.omto.2018.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/07/2018] [Indexed: 12/26/2022]
Abstract
Previous studies showed that the COpper Metabolism gene MURR1 Domain (COMMD) family of proteins was abnormally expressed in hepatocellular carcinoma (HCC). This study aimed to explore the roles of COMMD1 and COMMD7 in regulating nuclear factor κB (NF-κB) signaling in HCC stem cells (HCSCs). In vivo, the expression of COMMD7 and COMMD1 was determined in 35 pairs of HCC cancer tissues and adjacent tissues, and the effect of COMMD7 silencing on xenograft tumor growth was evaluated. In vitro, the effects of COMMD7 silencing and COMMD1 overexpression on HCSC function were assessed. Results found that the expression levels of COMMD7 were higher, whereas COMMD1 levels were lower in HCC tissues and HCSCs. COMMD7 silencing or COMMD1 overexpression inhibited cell proliferation, migration, and invasion through suppression of NF-κB p65. Furthermore, COMMD7 positively regulated NF-κB by upregulating protein inhibitor for activated stat 4 (PIAS4). This study demonstrates that COMMD7 has a dual regulatory role in the NF-κB signaling pathway in Nanog+ HCSCs.
Collapse
Affiliation(s)
- Lu Zheng
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Army Medical University, PLA, Chongqing, China
| | - Nan You
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Army Medical University, PLA, Chongqing, China
| | - Xiaobing Huang
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Army Medical University, PLA, Chongqing, China
| | - Huiying Gu
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Army Medical University, PLA, Chongqing, China
| | - Ke Wu
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Army Medical University, PLA, Chongqing, China
| | - Na Mi
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Army Medical University, PLA, Chongqing, China
| | - Jing Li
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Army Medical University, PLA, Chongqing, China
| |
Collapse
|
50
|
Podany A, Rauchut J, Wu T, Kawasawa YI, Wright J, Lamendella R, Soybel DI, Kelleher SL. Excess Dietary Zinc Intake in Neonatal Mice Causes Oxidative Stress and Alters Intestinal Host-Microbe Interactions. Mol Nutr Food Res 2018; 63:e1800947. [PMID: 30513548 DOI: 10.1002/mnfr.201800947] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/23/2018] [Indexed: 12/11/2022]
Abstract
SCOPE Greater than 68% of young infants are exposed to dietary zinc (Zn) levels that are higher than the Tolerable Upper Intake Limit. However, the consequences of excess dietary Zn during early life on intestinal function and host-microbe interactions are unknown. METHODS AND RESULTS Neonatal mice are gavaged with 100 Zn µg d-1 from postnatal day (PN) 2 through PN10 and indices of intestinal function and host-microbe interactions are compared to unsupplemented mice. Excess dietary Zn causes oxidative stress, increases goblet cell number and mucus production, and are associated with increased intestinal permeability and systemic inflammation. Over 900 genes are differentially expressed; 413 genes display a fold-change >1.60. The Gene Ontology Biological processes most significantly affected include biological adhesion, the immune system, metabolic processes, and response to stimulus. Key genes most highly and significantly upregulated include ALDH2, MT1, TMEM6, CDK20, and COX62b, while CALU, ST3GAL4, CRTC2, SLC28A2, and COMMA1 are downregulated. These changes are associated with a microbiome enriched in pathogenic taxa including Pseudomonadales and Campylobacter, and greater expression of bacterial stress response genes. CONCLUSION Excess dietary Zn may have unforeseen influences on epithelial signaling pathways, barrier function, and luminal ecology in the intestine that may have long-term consequences on intestinal health.
Collapse
Affiliation(s)
- Abigail Podany
- Department of Surgery, Penn State Hershey College of Medicine, Hershey, PA
| | - Jessica Rauchut
- Department of Surgery, Penn State Hershey College of Medicine, Hershey, PA
| | - Tong Wu
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Penn State Hershey College of Medicine, Hershey, PA.,Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, Penn State Hershey College of Medicine, Hershey, PA
| | - Justin Wright
- Department of Biology, Juniata College, Huntingdon, PA
| | | | - David I Soybel
- Department of Surgery, Penn State Hershey College of Medicine, Hershey, PA
| | - Shannon L Kelleher
- Department of Surgery, Penn State Hershey College of Medicine, Hershey, PA.,Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA.,Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, PA
| |
Collapse
|