1
|
Li L, Bu X, Wang S, Liu Y, Chen C, Zhang W, Zhao P. Response Gene to Complement 32 is associated with poor patient survival and an inflamed tumor-immune microenvironment in clear cell renal cell carcinoma. Transl Oncol 2025; 52:102248. [PMID: 39709718 PMCID: PMC11832949 DOI: 10.1016/j.tranon.2024.102248] [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: 05/15/2024] [Revised: 11/13/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024] Open
Abstract
It has been well established that tumor-infiltrating lymphocytes (TILs) play a critical role in the pathogenesis and progression of clear cell renal cell carcinoma (ccRCC). However, the mechanism on the interactions between TILs and tumor cells in the tumor-immune microenvironment remains unclear. In the present study, the expression of Response Gene to Complement 32 (RGC-32) was evaluated using immunohistochemistry. We analyzed the associations of RGC-32 expression with patient characteristics and survival. We also assessed TILs and their subsets (CD3+, CD4+, CD8+ and PD-1+) in the tumor nest. The level of RGC-32 expression was positively correlated with ISUP grade and Ki67 expression and was an independent poor prognosis factor of patients with ccRCC. RGC-32 expression was negatively correlated with the infiltration of TIL and CD3+T cells, but positively correlated with infiltration of PD-1+cells. In vitro studies showed that RGC-32 expression in renal cancer cells was downregulated by activated immune cells. Further investigation revealed that RGC-32 expression in renal cancer cells was inhibited by TNF-α and IL-1β secreted by activated immune cells. Collectively, these data indicate that RGC-32 could be a novel prognostic and druggable target related to the tumor-immune microenvironment in renal cancer.
Collapse
Affiliation(s)
- Lingling Li
- School of Basic Medicine, Shandong Second Medical University, Weifang, China
| | - Xiaocui Bu
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| | - Shuhui Wang
- Clinical laboratory, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yan Liu
- Department of Pathology, The 971 Hospital of People's Liberation Army Navy, Qingdao, China
| | - Chongdao Chen
- Department of Pathology, The 971 Hospital of People's Liberation Army Navy, Qingdao, China
| | - Wei Zhang
- Department of Pathology, The 971 Hospital of People's Liberation Army Navy, Qingdao, China.
| | - Peng Zhao
- Biotherapy Center, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
2
|
Tatomir A, Vlaicu S, Nguyen V, Luzina IG, Atamas SP, Drachenberg C, Papadimitriou J, Badea TC, Rus HG, Rus V. RGC-32 mediates proinflammatory and profibrotic pathways in immune-mediated kidney disease. Clin Immunol 2024; 265:110279. [PMID: 38878807 DOI: 10.1016/j.clim.2024.110279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Systemic lupus erythematosus is an autoimmune disease that results in immune-mediated damage to kidneys and other organs. We investigated the role of response gene to complement-32 (RGC-32), a proinflammatory and profibrotic mediator induced by TGFβ and C5b-9, in nephrotoxic nephritis (NTN), an experimental model that mimics human lupus nephritis. Proteinuria, loss of renal function and kidney histopathology were attenuated in RGC-32 KO NTN mice. RGC-32 KO NTN mice displayed downregulation of the CCL20/CCR6 and CXCL9/CXCR3 ligand/receptor pairs resulting in decreased renal recruitment of IL-17+ and IFNγ+ cells and subsequent decrease in the influx of innate immune cells. RGC-32 deficiency attenuated renal fibrosis as demonstrated by decreased deposition of collagen I, III and fibronectin. Thus, RGC-32 is a unique mediator shared by the Th17 and Th1 dependent proinflammatory and profibrotic pathways and a potential novel therapeutic target in the treatment of immune complex mediated glomerulonephritis such as lupus nephritis.
Collapse
Affiliation(s)
- Alexandru Tatomir
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA; Neurology Service, Veterans Administration Medical Health Care Center, Baltimore, MD, USA
| | - Sonia Vlaicu
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Internal Medicine, Medical Clinic nr. 1, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Vinh Nguyen
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Irina G Luzina
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sergei P Atamas
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | - Tudor C Badea
- Research and Development Institute, Faculty of Medicine, Transylvania University of Brasov, Brasov, Romania
| | - Horea G Rus
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA; Neurology Service, Veterans Administration Medical Health Care Center, Baltimore, MD, USA
| | - Violeta Rus
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
3
|
Mareco EA, de la Serrana DG, de Paula TG, Zanella BTT, da Silva Duran BO, Salomão RAS, de Almeida Fantinatti BE, de Oliveira VHG, Dos Santos VB, Carvalho RF, Dal-Pai-Silva M. Transcriptomic insight into the hybridization mechanism of the Tambacu, a hybrid from Colossoma macropomum (Tambaqui) and Piaractus mesopotamicus (Pacu). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2023; 45:101041. [PMID: 36442404 DOI: 10.1016/j.cbd.2022.101041] [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: 07/10/2022] [Revised: 11/02/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022]
Abstract
Interspecific hybrids are highly complex organisms, especially considering aspects related to the organization of genetic material. The diversity of possibilities created by the genetic combination between different species makes it difficult to establish a large-scale analysis methodology. An example of this complexity is Tambacu, an interspecific hybrid of Colossoma macropomum (Tambaqui) and Piaractus mesopotamicus (Pacu). Either genotype represents an essential role in South American aquaculture. However, despite this importance, the genetic information for these genotypes is still highly scarce in specialized databases. Using RNA-Seq analysis, we characterized the transcriptome of white muscle from Pacu, Tambaqui, and their interspecific hybrid (Tambacu). The sequencing process allowed us to obtain a significant number of reads (approximately 53 billion short reads). A total of annotated contigs were 37,285, 96,738, and 158,709 for Pacu, Tambaqui, and Tambacu. After that, we performed a comparative analysis of the transcriptome of the three genotypes, where we evaluated the differential expression (Tambacu vs Pacu = 11,156, and Tambacu vs Tambaqui = 876) profile of the transcript and the degree of similarity between the nucleotide sequences between the genotypes. We assessed the intensity and pattern of expression across genotypes using differential expression information. Clusterization analysis showed a closer relationship between Tambaqui and Tambacu. Furthermore, digital differential expression analysis selected some target genes related to essential cellular processes to evaluate and validate the expression through the RT-qPCR. The RT-qPCR analysis demonstrated significantly (p < 0.05) elevated expression of the mafbx, foxo1a, and rgcc genes in the hybrid compared to the parents. Likewise, we can observe genes significantly more expressed in Pacu (mtco1 and mylpfa) and mtco2 in Tambaqui. Our results showed that the phenotype presented by Tambacu might be associated with changes in the gene expression profile and not necessarily with an increase in gene variability. Thus, the molecular mechanisms underlying these "hybrid effects" may be related to additive and, in some cases, dominant regulatory interactions between parental alleles that act directly on gene regulation in the hybrid transcripts.
Collapse
Affiliation(s)
- Edson Assunção Mareco
- Environment and Regional Development Graduate Program, University of Western São Paulo, Presidente Prudente, São Paulo, Brazil; Biology Department, University of Western São Paulo, Presidente Prudente, São Paulo, Brazil.
| | - Daniel Garcia de la Serrana
- Cell Biology, Physiology, and Immunology Department, School of Biology, University of Barcelona, 643 08028 Barcelona, Catalonia, Spain
| | - Tassiana Gutierrez de Paula
- Department of Structural and Functional Biology, Institute of Bioscience of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Bruna Tereza Thomazini Zanella
- Department of Structural and Functional Biology, Institute of Bioscience of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Bruno Oliveira da Silva Duran
- Department of Structural and Functional Biology, Institute of Bioscience of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | | | | | - Victor Hugo Garcia de Oliveira
- Environment and Regional Development Graduate Program, University of Western São Paulo, Presidente Prudente, São Paulo, Brazil
| | | | - Robson Francisco Carvalho
- Department of Structural and Functional Biology, Institute of Bioscience of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Maeli Dal-Pai-Silva
- Department of Structural and Functional Biology, Institute of Bioscience of Botucatu, São Paulo State University, Botucatu, São Paulo, Brazil
| |
Collapse
|
4
|
Luzina IG, Rus V, Lockatell V, Courneya JP, Hampton BS, Fishelevich R, Misharin AV, Todd NW, Badea TC, Rus H, Atamas SP. Regulator of Cell Cycle Protein (RGCC/RGC-32) Protects against Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2022; 66:146-157. [PMID: 34668840 PMCID: PMC8845131 DOI: 10.1165/rcmb.2021-0022oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Some previous studies in tissue fibrosis have suggested a profibrotic contribution from elevated expression of a protein termed either RGCC (regulator of cell cycle) or RGC-32 (response gene to complement 32 protein). Our analysis of public gene expression datasets, by contrast, revealed a consistent decrease in RGCC mRNA levels in association with pulmonary fibrosis. Consistent with this observation, we found that stimulating primary adult human lung fibroblasts with transforming growth factor (TGF)-β in cell cultures elevated collagen expression and simultaneously attenuated RGCC mRNA and protein levels. Moreover, overexpression of RGCC in cultured lung fibroblasts attenuated the stimulating effect of TGF-β on collagen levels. Similar to humans with pulmonary fibrosis, the levels of RGCC were also decreased in vivo in lung tissues of wild-type mice challenged with bleomycin in both acute and chronic models. Mice with constitutive RGCC gene deletion accumulated more collagen in their lungs in response to chronic bleomycin challenge than did wild-type mice. RNA-Seq analyses of lung fibroblasts revealed that RGCC overexpression alone had a modest transcriptomic effect, but in combination with TGF-β stimulation, induced notable transcriptomic changes that negated the effects of TGF-β, including on extracellular matrix-related genes. At the level of intracellular signaling, RGCC overexpression delayed early TGF-β-induced Smad2/3 phosphorylation, elevated the expression of total and phosphorylated antifibrotic mediator STAT1, and attenuated the expression of a profibrotic mediator STAT3. We conclude that RGCC plays a protective role in pulmonary fibrosis and that its decline permits collagen accumulation. Restoration of RGCC expression may have therapeutic potential in pulmonary fibrosis.
Collapse
Affiliation(s)
- Irina G. Luzina
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Violeta Rus
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Virginia Lockatell
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Jean-Paul Courneya
- Health Sciences and Human Services Library, University of Maryland–Baltimore, Baltimore, Maryland
| | | | - Rita Fishelevich
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Alexander V. Misharin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University, Chicago, Illinois
| | - Nevins W. Todd
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Tudor C. Badea
- Retinal Circuits Development and Genetics Unit, National Eye Institute, Bethesda, Maryland; and,Faculty of Medicine, Research and Development Institute, Transilvania University of Brașov, Brașov, Romania
| | - Horea Rus
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| | - Sergei P. Atamas
- University of Maryland School of Medicine, Baltimore, Maryland;,Baltimore VA Medical Center, Baltimore, Maryland
| |
Collapse
|
5
|
Hanoudi SN, Talwar H, Draghici S, Samavati L. Autoantibodies against cytoskeletons and lysosomal trafficking discriminate sarcoidosis from healthy controls, tuberculosis and lung cancers. MOLECULAR BIOMEDICINE 2022; 3:3. [PMID: 35048206 PMCID: PMC8770712 DOI: 10.1186/s43556-021-00064-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
Sarcoidosis is a systemic granulomatous disease of unknown etiology. Hypergammaglobulinemia and the presence of autoantibodies in sarcoidosis suggest active humoral immunity to unknown antigen(s). We developed a complex cDNA library derived from tissues of sarcoidosis patients. Using a high throughput method, we constructed a microarray platform from this cDNA library containing large numbers of sarcoidosis clones. After selective biopanning, 1070 sarcoidosis-specifc clones were arrayed and immunoscreend with 152 sera from patients with sarcoidosis and other pulmonary diseases. To identify the sarcoidosis classifiers two statistical approaches were conducted: First, we identified significant biomarkers between sarcoidosis and healthy controls, and second identified markers comparing sarcoidosis to all other groups. At the threshold of an False Discovery Rate (FDR) < 0.01, we identified 14 clones in the first approach and 12 clones in the second approach discriminating sarcoidosis from other groups. We used the classifiers to build a naïve Bayes model on the training-set and validated it on an independent test-set. The first approach yielded an AUC of 0.947 using 14 significant clones with a sensitivity of 0.93 and specificity of 0.88, whereas the AUC of the second option was 0.92 with a sensitivity of 0.96 and specificity of 0.83. These results suggest robust classifier performance. Furthermore, we characterized the informative phage clones by sequencing and homology searches. Large numbers of classifier-clones were peptides involved in cellular trafficking and cytoskeletons. These results show that sarcoidosis is associated with a specific pattern of immunoreactivity that can discriminate it from other diseases.
Collapse
Affiliation(s)
| | - Harvinder Talwar
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine, 3990 John R, 3 Hudson, Detroit, MI 48201 USA
| | - Sorin Draghici
- Department of Computer Science, Wayne State University, Detroit, MI 48202 USA
| | - Lobelia Samavati
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine, 3990 John R, 3 Hudson, Detroit, MI 48201 USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, 540 E. Canfield, Detroit, MI 48201 USA
| |
Collapse
|
6
|
Jin M, Lu J, Fei X, Lu Z, Quan K, Liu Y, Chu M, Di R, Wei C, Wang H. Selection Signatures Analysis Reveals Genes Associated with High-Altitude Adaptation in Tibetan Goats from Nagqu, Tibet. Animals (Basel) 2020; 10:ani10091599. [PMID: 32911823 PMCID: PMC7552128 DOI: 10.3390/ani10091599] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/28/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary In the process of domestication, goats have undergone long-term artificial and natural selection, leading to differences among goat breeds and leaving different selection traces on the genome. However, the genetic components underlying high-altitude adaptation remain largely unknown. Here, we genotyped four goat breeds using the Illumina Caprine 50K single nucleotide polymorphism (SNP) Chip. One highland breed (Tibetan goat) compared with three lowland breeds (Huanghuai goat, Taihang goat and Xinjiang goat) to identify the molecular basis of high-altitude adaptation. So, we investigated selection signatures using the di statistic of four goat breeds and some genes in Tibetan goats related to high-altitude adaptation were identified. In addition, q-PCR validated the gene expression level in Tibetan goats and Huanghuai goats. This information may be valuable for the study of the genetic uniqueness of Tibetan goats and increased understanding of the hypoxic adaptation mechanism of Tibetan goats on the plateau. Abstract Tibetan goat is an ancient breed, which inhabits the adverse conditions of the plateaus in China. To investigate the role of selection in shaping its genomes, we genotyped Tibetan goats (Nagqu Prefecture, above 4500 m) and three lowland populations (Xinjiang goats, Taihang goats and Huanghuai goats). The result of PCA, neighbor-joining (N-J) tree and model-based clustering showed that the genetic structure between the Tibetan goat and the three lowland populations has significant difference. As demonstrated by the di statistic, we found that some genes were related to the high-altitude adaptation of Tibetan goats. Functional analysis revealed that these genes were enriched in the VEGF (vascular endothelial growth factor) signaling pathway and melanoma, suggesting that nine genes (FGF2, EGFR, AKT1, PTEN, MITF, ENPEP, SIRT6, KDR, and CDC42) might have important roles in the high-altitude adaptation of Nagqu Tibetan goats. We also found that the LEPR gene was under the strongest selection (di value = 16.70), and it could induce upregulation of the hypoxic ventilatory response. In addition, five genes (LEPR, LDB1, EGFR, NOX4 and FGF2) with high di values were analyzed using q-PCR. Among them, we found that LEPR, LDB1 and FGF2 exhibited higher expression in the lungs of the Tibetan goats; LEPR, EGFR and LDB1 exhibited higher expression in the hearts of the Huanghuai goat. Our results suggest that LEPR, LDB1, EGFR and FGF2 genes may be related to the high-altitude adaptation of the goats. These findings improve our understanding of the selection of the high-altitude adaptability of the Nagqu Tibetan goats and provide new theoretical knowledge for the conservation and utilization of germplasm resources.
Collapse
Affiliation(s)
- Meilin Jin
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
| | - Jian Lu
- National Animal Husbandry Service, Beijing 100193, China;
| | - Xiaojuan Fei
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
| | - Zengkui Lu
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China;
| | - Kai Quan
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China;
| | - Yongbin Liu
- Inner Mongolia Academy of Animal Husbandry Science, Hohhot 010031, China;
| | - Mingxing Chu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
| | - Ran Di
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
| | - Caihong Wei
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
- Correspondence: (C.W.); (H.W.)
| | - Huihua Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
- Correspondence: (C.W.); (H.W.)
| |
Collapse
|
7
|
Liu R, Xu B, Yu S, Zhang J, Sun H, Liu C, Lu F, Pan Q, Zhang X. Integrated Transcriptomic and Proteomic Analyses of the Interaction Between Chicken Synovial Fibroblasts and Mycoplasma synoviae. Front Microbiol 2020; 11:576. [PMID: 32318048 PMCID: PMC7147270 DOI: 10.3389/fmicb.2020.00576] [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: 12/05/2019] [Accepted: 03/16/2020] [Indexed: 12/29/2022] Open
Abstract
Mycoplasma synoviae (MS), which causes respiratory disease, eggshell apex abnormalities, infectious synovitis, and arthritis in avian species, has become an economically detrimental poultry pathogen in recent years. In China, the disease is characterized by infectious synovitis and arthritis. However, the mechanism by which MS causes infectious synovitis and arthritis remains unknown. Increasing evidence suggests that synovial fibroblasts (SF) play a key role in the pathogenesis of arthritis. Here, both RNA sequencing and tandem mass tag analyses are utilized to compare the response of primary chicken SF (CSF) following infection with and without MS. The host response between non-infected and infected cells was remarkably different at both the mRNA and protein levels. In total, 2,347 differentially expressed genes (DEGs) (upregulated, n = 1,137; downregulated, n = 1,210) and 221 differentially expressed proteins (DEPs) (upregulated, n = 129; downregulated, n = 92) were detected in the infected group. A correlation analysis indicated a moderate positive correlation between the mRNA and protein level changes in MS-infected CSF. At both the transcriptomic and proteomic levels, 149 DEGs were identified; 88 genes were upregulated and 61 genes were downregulated in CSF. Additionally, part of these regulated genes and their protein products were grouped into seven categories: proliferation-related and apoptosis-related factors, inflammatory mediators, proangiogenic factors, antiangiogenic factors, matrix metalloproteinases, and other arthritis-related proteins. These proteins may be involved in the pathogenesis of MS-induced arthritis in chickens. To our knowledge, this is the first integrated analysis on the mechanism of CSF-MS interactions that combined transcriptomic and proteomic technologies. In this study, many key candidate genes and their protein products related to MS-induced infectious synovitis and arthritis were identified.
Collapse
Affiliation(s)
- Rui Liu
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China.,National Center for Engineering Research of Veterinary Bio-products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Bin Xu
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China.,National Center for Engineering Research of Veterinary Bio-products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Shengqing Yu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Jingfeng Zhang
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China.,National Center for Engineering Research of Veterinary Bio-products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Huawei Sun
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China.,National Center for Engineering Research of Veterinary Bio-products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Chuanmin Liu
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China.,National Center for Engineering Research of Veterinary Bio-products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Fengying Lu
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China.,National Center for Engineering Research of Veterinary Bio-products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Qunxing Pan
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China.,National Center for Engineering Research of Veterinary Bio-products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Xiaofei Zhang
- Key Laboratory of Veterinary Biological Engineering and Technology, Ministry of Agriculture, Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China.,National Center for Engineering Research of Veterinary Bio-products, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| |
Collapse
|
8
|
Single-cell transcriptomics of the human retinal pigment epithelium and choroid in health and macular degeneration. Proc Natl Acad Sci U S A 2019; 116:24100-24107. [PMID: 31712411 PMCID: PMC6883845 DOI: 10.1073/pnas.1914143116] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The retinal pigment epithelium and the choroid are complex tissues whose dysfunction can lead to irreversible visual loss. In this study, single-cell RNA sequencing of both of these tissues was performed to characterize gene expression patterns specific to the retinal pigment epithelium and all major choroidal cell populations. Unique gene expression signatures of arterial, venous, and choriocapillaris vascular beds within the choroid were identified. RGCC, a gene that responds to complement and has been shown to induce endothelial apoptosis, was specifically expressed in choriocapillaris endothelial cells. This study provides potential insight into the molecular mechanisms of choroidal vascular disease and its contribution to age-related macular degeneration. The human retinal pigment epithelium (RPE) and choroid are complex tissues that provide crucial support to the retina. Disease affecting either of these supportive tissues can lead to irreversible blindness in the setting of age-related macular degeneration. In this study, single-cell RNA sequencing was performed on macular and peripheral regions of RPE-choroid from 7 human donor eyes in 2 independent experiments. In the first experiment, total RPE/choroid preparations were evaluated and expression profiles specific to RPE and major choroidal cell populations were identified. As choroidal endothelial cells represent a minority of the total RPE/choroidal cell population but are strongly implicated in age-related macular degeneration (AMD) pathogenesis, a second single-cell RNA-sequencing experiment was performed using endothelial cells enriched by magnetic separation. In this second study, we identified gene expression signatures along the choroidal vascular tree, classifying the transcriptome of human choriocapillaris, arterial, and venous endothelial cells. We found that the choriocapillaris highly and specifically expresses the regulator of cell cycle gene (RGCC), a gene that responds to complement activation and induces apoptosis in endothelial cells. In addition, RGCC was the most up-regulated choriocapillaris gene in a donor diagnosed with AMD. These results provide a characterization of the human RPE and choriocapillaris transcriptome, offering potential insight into the mechanisms of choriocapillaris response to complement injury and choroidal vascular disease in age-related macular degeneration.
Collapse
|
9
|
Ma B, Li M, Fuchs S, Bischoff I, Hofmann A, Unger RE, Kirkpatrick CJ. Short‐term hypoxia promotes vascularization in co‐culture system consisting of primary human osteoblasts and outgrowth endothelial cells. J Biomed Mater Res A 2019; 108:7-18. [DOI: 10.1002/jbm.a.36786] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 01/23/2023]
Affiliation(s)
- Bin Ma
- Institute of Pathology, University Medical Centre of the Johannes Gutenberg University Mainz Mainz Germany
- Medical, Molecular and Forensic SciencesMurdoch University Murdoch Western Australia Australia
| | - Ming Li
- Institute of Pathology, University Medical Centre of the Johannes Gutenberg University Mainz Mainz Germany
| | - Sabine Fuchs
- Institute of Pathology, University Medical Centre of the Johannes Gutenberg University Mainz Mainz Germany
- Experimental Trauma SurgeryUniversity Medical Center Schleswig‐Holstein Kiel Kiel Germany
| | - Iris Bischoff
- Institute of Pathology, University Medical Centre of the Johannes Gutenberg University Mainz Mainz Germany
| | - Alexander Hofmann
- Department of Trauma SurgeryUniversity Medical Centre of the Johannes Gutenberg University Mainz Mainz Germany
| | - Ronald E. Unger
- Institute of Pathology, University Medical Centre of the Johannes Gutenberg University Mainz Mainz Germany
| | - Charles J. Kirkpatrick
- Institute of Pathology, University Medical Centre of the Johannes Gutenberg University Mainz Mainz Germany
| |
Collapse
|
10
|
Vlaicu SI, Tatomir A, Rus V, Rus H. Role of C5b-9 and RGC-32 in Cancer. Front Immunol 2019; 10:1054. [PMID: 31156630 PMCID: PMC6530392 DOI: 10.3389/fimmu.2019.01054] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/24/2019] [Indexed: 01/13/2023] Open
Abstract
The complement system represents an effective arsenal of innate immunity as well as an interface between innate and adaptive immunity. Activation of the complement system culminates with the assembly of the C5b-9 terminal complement complex on cell membranes, inducing target cell lysis. Translation of this sequence of events into a malignant setting has traditionally afforded C5b-9 a strict antitumoral role, in synergy with antibody-dependent tumor cytolysis. However, in recent decades, a plethora of evidence has revised this view, highlighting the tumor-promoting properties of C5b-9. Sublytic C5b-9 induces cell cycle progression by activating signal transduction pathways (e.g., Gi protein/ phosphatidylinositol 3-kinase (PI3K)/Akt kinase and Ras/Raf1/ERK1) and modulating the activation of cancer-related transcription factors, while shielding malignant cells from apoptosis. C5b-9 also induces Response Gene to Complement (RGC)-32, a gene that contributes to cell cycle regulation by activating the Akt and CDC2 kinases. RGC-32 is expressed by tumor cells and plays a dual role in cancer, functioning as either a tumor promoter by endorsing malignancy initiation, progression, invasion, metastasis, and angiogenesis, or as a tumor suppressor. In this review, we present recent data describing the versatile, multifaceted roles of C5b-9 and its effector, RGC-32, in cancer.
Collapse
Affiliation(s)
- Sonia I Vlaicu
- Department of Internal Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Neurology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Alexandru Tatomir
- Department of Neurology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Violeta Rus
- Division of Rheumatology and Immunology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Horea Rus
- Department of Neurology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
11
|
Tanase-Nakao K, Mizuno K, Hayashi Y, Kojima Y, Hara M, Matsumoto K, Matsubara Y, Igarashi M, Miyado M, Fukami M. Dihydrotestosterone induces minor transcriptional alterations in genital skin fibroblasts of children with and without androgen insensitivity. Endocr J 2019; 66:387-393. [PMID: 30787207 DOI: 10.1507/endocrj.ej18-0494] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Endogenous and exogenous androgens induce masculinization of external genitalia through binding to the androgen receptor (AR). The target genes of androgens in external genitalia remain to be determined, although previous studies have shown that the apolipoprotein D gene (APOD) was significantly upregulated by dihydrotestosterone (DHT), the most potent androgen in humans. In the present study, we performed microarray analysis for genital skin fibroblasts obtained from four boys with buried penis (the control individuals) and a patient with partial androgen insensitivity syndrome (PAIS) due to a hypomorphic mutation in AR (the PAIS patient). We identified 24 transcripts that were upregulated or downregulated by DHT in all samples of control individuals and, to a lesser extent, in the sample of the PAIS patient. Differences between DHT-treated and -untreated samples were small; the results of 24 transcripts did not reach statistical significance. The 24 transcripts included CYP1B1, a gene possibly involved in the development of genital tubercle in mice, and APOD, as well as several genes that have been reported as androgen targets in prostate or other tissues. The results of this study indicate that androgen-mediated masculinization of external genitalia is unlikely to depend on massive transcriptional changes in specific AR target genes. Rather, minor transcriptional changes of several genes, and/or a complex molecular network may play a major role in penile development. Importantly, our data suggest the possible involvement of CYP1B1 in human genital development and confirm the clinical importance of APOD as a biomarker for AR function.
Collapse
Affiliation(s)
- Kanako Tanase-Nakao
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
- Department of Advanced Pediatric Medicine, Tohoku University School of Medicine, Tokyo 157-8535, Japan
| | - Kentaro Mizuno
- Department of Pediatric Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Yutaro Hayashi
- Department of Pediatric Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Yoshiyuki Kojima
- Department of Urology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Mariko Hara
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Yoichi Matsubara
- Department of Advanced Pediatric Medicine, Tohoku University School of Medicine, Tokyo 157-8535, Japan
- Institute director, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Maki Igarashi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Mami Miyado
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
12
|
Liao WL, Lin JM, Liu SP, Chen SY, Lin HJ, Wang YH, Lei YJ, Huang YC, Tsai FJ. Loss of Response Gene to Complement 32 (RGC-32) in Diabetic Mouse Retina Is Involved in Retinopathy Development. Int J Mol Sci 2018; 19:ijms19113629. [PMID: 30453650 PMCID: PMC6275084 DOI: 10.3390/ijms19113629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/06/2018] [Accepted: 11/15/2018] [Indexed: 12/14/2022] Open
Abstract
Diabetic retinopathy (DR) is a severe and recurrent microvascular complication in diabetes. The multifunctional response gene to complement 32 (RGC-32) is involved in the regulation of cell cycle, proliferation, and apoptosis. To investigate the role of RGC-32 in the development of DR, we used human retinal microvascular endothelial cells under high-glucose conditions and type 2 diabetes (T2D) mice (+Leprdb/ + Leprdb, db/db). The results showed that RGC-32 expression increased moderately in human retinal endothelial cells under hyperglycemic conditions. Histopathology and RGC-32 expression showed no significant changes between T2D and control mice retina at 16 and 24 weeks of age. However, RGC-32 expression was significantly decreased in T2D mouse retina compared to the control group at 32 weeks of age, which develop features of the early clinical stages of DR, namely reduced retinal thickness and increased ganglion cell death. Moreover, immunohistochemistry showed that RGC-32 was predominantly expressed in the photoreceptor inner segments of control mice, while the expression was dramatically lowered in the T2D retinas. Furthermore, we found that the level of anti-apoptotic protein Bcl-2 was decreased (approximately 2-fold) with a concomitant increase in cleaved caspase-3 (approximately 3-fold) in T2D retina compared to control. In summary, RGC-32 may lose its expression in T2D retina with features of DR, suggesting that it plays a critical role in DR pathogenesis.
Collapse
Affiliation(s)
- Wen-Ling Liao
- Center for Personalized Medicine, China Medical University Hospital and Graduate Institute of Integrated Medicine, China Medical University, Taichung 404, Taiwan.
| | - Jane-Ming Lin
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
- Department of Ophthalmology, China Medical University Hospital, Taichung 404, Taiwan.
| | - Shih-Ping Liu
- Center for Translational Medicine, China Medical University Hospital and Graduate Institute of Biomedical Science, China Medical University, Taichung 404, Taiwan and Department of Social Work, Asia University, Taichung 413, Taiwan.
| | - Shih-Yin Chen
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan.
| | - Hui-Ju Lin
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
- Department of Ophthalmology, China Medical University Hospital, Taichung 404, Taiwan.
| | - Yeh-Han Wang
- Department of Anatomical Pathology, Taipei Institute of Pathology, Taipei 103, Taiwan and Institute of Public Health, National Yang-Ming University, Taipei 112, Taiwan.
| | - Yu-Jie Lei
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan.
| | - Yu-Chuen Huang
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan.
| | - Fuu-Jen Tsai
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
- Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan.
- Department of Medical Genetics, China Medical University Hospital and Children's Hospital of China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
13
|
Cui XB, Chen SY. Response Gene to Complement 32 in Vascular Diseases. Front Cardiovasc Med 2018; 5:128. [PMID: 30280101 PMCID: PMC6153333 DOI: 10.3389/fcvm.2018.00128] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/28/2018] [Indexed: 11/16/2022] Open
Abstract
Response gene to complement 32 (RGC32) is a protein that was identified in rat oligodendrocytes after complement activation. It is expressed in most of the organs and tissues, such as brain, placenta, heart, and the liver. Functionally, RGC32 is involved in various physiological and pathological processes, including cell proliferation, differentiation, fibrosis, metabolic disease, and cancer. Emerging evidences support the roles of RGC32 in vascular diseases. RGC32 promotes injury-induced vascular neointima formation by mediating smooth muscle cell (SMC) proliferation and migration. Moreover, RGC32 mediates endothelial cell activation and facilitates atherosclerosis development. Its involvement in macrophage phagocytosis and activation as well as T-lymphocyte cell cycle activation also suggests that RGC32 is important for the development and progression of inflammatory vascular diseases. In this mini-review, we provide an overview on the roles of RGC32 in regulating functions of SMCs, endothelial cells, and immune cells, and discuss their contributions to vascular diseases.
Collapse
Affiliation(s)
- Xiao-Bing Cui
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, United States
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA, United States
| |
Collapse
|
14
|
Chen YJ, Chang WA, Wu LY, Hsu YL, Chen CH, Kuo PL. Systematic Analysis of Differential Expression Profile in Rheumatoid Arthritis Chondrocytes Using Next-Generation Sequencing and Bioinformatics Approaches. Int J Med Sci 2018; 15:1129-1142. [PMID: 30123050 PMCID: PMC6097257 DOI: 10.7150/ijms.27056] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/08/2018] [Indexed: 12/15/2022] Open
Abstract
Cartilage destruction in rheumatoid arthritis (RA) occurs primarily in the pannus-cartilage interface. The close contact of the synovium-cartilage interface implicates crosstalk between synovial fibroblasts and chondrocytes. The aim of this study is to explore the differentially expressed genes and novel microRNA regulations potentially implicated in the dysregulated cartilage homeostasis in joint destruction of RA. Total RNAs were extracted from human primary cultured normal and RA chondrocytes for RNA and small RNA expression profiling using next-generation sequencing. Using systematic bioinformatics analyses, we identified 463 differentially expressed genes in RA chondrocytes were enriched in biological functions related to altered cell cycle process, inflammatory response and hypoxic stimulation. Moreover, fibroblast growth factor 9 (FGF9), kynureninase (KYNU), and regulator of cell cycle (RGCC) were among the top dysregulated genes identified to be potentially affected in the RA joint microenvironment, having similar expression patterns observed in arrays of clinical RA synovial tissues from the Gene Expression Omnibus database. Additionally, among the 31 differentially expressed microRNAs and 10 candidate genes with potential microRNA-mRNA interactions in RA chondrocytes, the novel miR-140-3p-FGF9 interaction was validated in different microRNA prediction databases, and proposed to participate in the pathogenesis of joint destruction through dysregulated cell growth in RA. The findings provide new perspectives for target genes in the management of cartilage destruction in RA.
Collapse
Affiliation(s)
- Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Wei-An Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ling-Yu Wu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chia-Hsin Chen
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
15
|
Woods L, Perez-Garcia V, Hemberger M. Regulation of Placental Development and Its Impact on Fetal Growth-New Insights From Mouse Models. Front Endocrinol (Lausanne) 2018; 9:570. [PMID: 30319550 PMCID: PMC6170611 DOI: 10.3389/fendo.2018.00570] [Citation(s) in RCA: 275] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/06/2018] [Indexed: 01/01/2023] Open
Abstract
The placenta is the chief regulator of nutrient supply to the growing embryo during gestation. As such, adequate placental function is instrumental for developmental progression throughout intrauterine development. One of the most common complications during pregnancy is insufficient growth of the fetus, a problem termed intrauterine growth restriction (IUGR) that is most frequently rooted in a malfunctional placenta. Together with conventional gene targeting approaches, recent advances in screening mouse mutants for placental defects, combined with the ability to rapidly induce mutations in vitro and in vivo by CRISPR-Cas9 technology, has provided new insights into the contribution of the genome to normal placental development. Most importantly, these data have demonstrated that far more genes are required for normal placentation than previously appreciated. Here, we provide a summary of common types of placental defects in established mouse mutants, which will help us gain a better understanding of the genes impacting on human placentation. Based on a recent mouse mutant screen, we then provide examples on how these data can be mined to identify novel molecular hubs that may be critical for placental development. Given the close association between placental defects and abnormal cardiovascular and brain development, these functional nodes may also shed light onto the etiology of birth defects that co-occur with placental malformations. Taken together, recent insights into the regulation of mouse placental development have opened up new avenues for research that will promote the study of human pregnancy conditions, notably those based on defects in placentation that underlie the most common pregnancy pathologies such as IUGR and pre-eclampsia.
Collapse
Affiliation(s)
- Laura Woods
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Vicente Perez-Garcia
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Vicente Perez-Garcia
| | - Myriam Hemberger
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
- Myriam Hemberger
| |
Collapse
|
16
|
Zhu L, Zhao Q. Hypoxia-inducible factor 1α participates in hypoxia-induced epithelial-mesenchymal transition via response gene to complement 32. Exp Ther Med 2017; 14:1825-1831. [PMID: 28810656 DOI: 10.3892/etm.2017.4665] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 04/10/2017] [Indexed: 12/11/2022] Open
Abstract
The aim of the present study was to explore the function of response gene to complement 32 (RGC-32) in hypoxia-induced epithelial-mesenchymal transition (EMT) in pancreatic cancer. Three kinds of hypoxia-inducible factor 1α (HIF-1α) small interfering (si)RNA were synthesized and the different effects on the expression of HIF-1α were detected by western blotting. In human pancreatic cancer BxPC-3 cells, HIF-1α levels were diminished using siRNA transfection or HIF-1α inhibitor pretreatment, and the expression levels of RGC-32 and EMT-associated proteins were analyzed using reverse transcription-quantitative polymerase chain reaction and western blotting. Subsequently, the protein levels of epithelial marker, E-cadherin, and mesenchymal marker, vimentin, were determined by western blotting. Results demonstrated that HIF-1α-Homo-488 siRNA and HIF-1α-Homo-1216 siRNA diminished the protein level of HIF-1α. Compared with normoxia, hypoxia induced the levels of HIF-1α, RGC-32, N-cadherin and vimentin, but suppressed the expression of E-cadherin and cytokeratins. The inhibition of HIF-1α by HIF-1α-Homo-1216 siRNA transfection or HIF-1α inhibitor repressed hypoxia-induced HIF-1α, RGC-32, N-cadherin and vimentin, but increased the expression of E-cadherin and cytokeratins. When RGC-32 was knocked down, hypoxia-induced vimentin was suppressed; however, hypoxia-suppressed N-cadherin was released. In conclusion, the present results demonstrated that hypoxia induced the expression of HIF-1α to activate the levels of RGC-32, in turn to regulate the expression EMT-associated proteins for EMT. These findings revealed the function of RGC-32 in hypoxia-induced EMT and may have identified a novel link between HIF-1α and EMT for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Liang Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
17
|
Counts SE, Mufson EJ. Regulator of Cell Cycle (RGCC) Expression During the Progression of Alzheimer's Disease. Cell Transplant 2016; 26:693-702. [PMID: 27938491 DOI: 10.3727/096368916x694184] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Unscheduled cell cycle reentry of postmitotic neurons has been described in cases of mild cognitive impairment (MCI) and Alzheimer's disease (AD) and may form a basis for selective neuronal vulnerability during disease progression. In this regard, the multifunctional protein regulator of cell cycle (RGCC) has been implicated in driving G1/S and G2/M phase transitions through its interactions with cdc/cyclin-dependent kinase 1 (cdk1) and is induced by p53, which mediates apoptosis in neurons. We tested whether RGCC levels were dysregulated in frontal cortex samples obtained postmortem from subjects who died with a clinical diagnosis of no cognitive impairment (NCI), MCI, or AD. RGCC mRNA and protein levels were upregulated by ∼50%-60% in MCI and AD compared to NCI, and RGCC protein levels were associated with poorer antemortem global cognitive performance in the subjects examined. To test whether RGCC might regulate neuronal cell cycle reentry and apoptosis, we differentiated neuronotypic PC12 cultures with nerve growth factor (NGF) followed by NGF withdrawal to induce abortive cell cycle activation and cell death. Experimental reduction of RGCC levels increased cell survival and reduced levels of the cdk1 target cyclin B1. RGCC may be a candidate cell cycle target for neuroprotection during the onset of AD.
Collapse
|
18
|
Vlaicu SI, Tatomir A, Boodhoo D, Ito T, Fosbrink M, Cudrici C, Mekala AP, Ciriello J, Crişan D, Boţan E, Rus V, Rus H. RGC-32 is expressed in the human atherosclerotic arterial wall: Role in C5b-9-induced cell proliferation and migration. Exp Mol Pathol 2016; 101:221-230. [DOI: 10.1016/j.yexmp.2016.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/07/2016] [Indexed: 01/21/2023]
|
19
|
Promoter Methylation and mRNA Expression of Response Gene to Complement 32 in Breast Carcinoma. J Cancer Epidemiol 2016; 2016:7680523. [PMID: 27118972 PMCID: PMC4828546 DOI: 10.1155/2016/7680523] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/23/2015] [Accepted: 03/08/2016] [Indexed: 12/21/2022] Open
Abstract
Background. Response gene to complement 32 (RGC32), induced by activation of complements, has been characterized as a cell cycle regulator; however, its role in carcinogenesis is still controversial. In the present study we compared RGC32 promoter methylation patterns and mRNA expression in breast cancerous tissues and adjacent normal tissues. Materials and Methods. Sixty-three breast cancer tissues and 63 adjacent nonneoplastic tissues were included in our study. Design. Nested methylation-specific polymerase chain reaction (Nested-MSP) and quantitative PCR (qPCR) were used to determine RGC32 promoter methylation status and its mRNA expression levels, respectively. Results. RGC32 methylation pattern was not different between breast cancerous tissue and adjacent nonneoplastic tissue (OR = 2.30, 95% CI = 0.95–5.54). However, qPCR analysis displayed higher levels of RGC32 mRNA in breast cancerous tissues than in noncancerous tissues (1.073 versus 0.959; P = 0.001), irrespective of the promoter methylation status. The expression levels and promoter methylation of RGC32 were not correlated with any of patients' clinical characteristics (P > 0.05). Conclusion. Our findings confirmed upregulation of RGC32 in breast cancerous tumors, but it was not associated with promoter methylation patterns.
Collapse
|
20
|
Zhao P, Gao D, Wang Q, Song B, Shao Q, Sun J, Ji C, Li X, Li P, Qu X. Response gene to complement 32 (RGC-32) expression on M2-polarized and tumor-associated macrophages is M-CSF-dependent and enhanced by tumor-derived IL-4. Cell Mol Immunol 2015; 12:692-9. [PMID: 25418473 PMCID: PMC4716617 DOI: 10.1038/cmi.2014.108] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 10/01/2014] [Accepted: 10/01/2014] [Indexed: 02/08/2023] Open
Abstract
Response gene to complement 32 (RGC-32) is a cell cycle regulator involved in the proliferation, differentiation and migration of cells and has also been implicated in angiogenesis. Here we show that RGC-32 expression in macrophages is induced by IL-4 and reduced by LPS, indicating a link between RGC-32 expression and M2 polarization. We demonstrated that the increased expression of RGC-32 is characteristic of alternatively activated macrophages, in which this protein suppresses the production of pro-inflammatory cytokine IL-6 and promotes the production of the anti-inflammatory mediator TGF-β. Consistent with in vitro data, tumor-associated macrophages (TAMs) express high levels of RGC-32, and this expression is induced by tumor-derived ascitic fluid in an M-CSF- and/or IL-4-dependent manner. Collectively, these results establish RGC-32 as a marker for M2 macrophage polarization and indicate that this protein is a potential target for cancer immunotherapy, targeting tumor-associated macrophages.
Collapse
Affiliation(s)
- Peng Zhao
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, China
- Biotherapy Center, Qingdao Central Hospital, the Second Affiliated Hospital, Qingdao University Medical College, Qingdao, China
| | - Daiqing Gao
- Biotherapy Center, Qingdao Central Hospital, the Second Affiliated Hospital, Qingdao University Medical College, Qingdao, China
| | - Qingjie Wang
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, China
| | - Bingfeng Song
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, China
| | - Qianqian Shao
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, China
| | - Jintang Sun
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, China
| | - Chunyan Ji
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, China
| | - Xingang Li
- Neurosurgery, Qilu Hospital of Shandong University, Jinan, China
| | - Peng Li
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, China
| | - Xun Qu
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
21
|
Vlaicu SI, Tatomir A, Rus V, Mekala AP, Mircea PA, Niculescu F, Rus H. The role of complement activation in atherogenesis: the first 40 years. Immunol Res 2015; 64:1-13. [DOI: 10.1007/s12026-015-8669-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
22
|
Tegla CA, Cudrici CD, Nguyen V, Danoff J, Kruszewski AM, Boodhoo D, Mekala AP, Vlaicu SI, Chen C, Rus V, Badea TC, Rus H. RGC-32 is a novel regulator of the T-lymphocyte cell cycle. Exp Mol Pathol 2015; 98:328-37. [PMID: 25770350 DOI: 10.1016/j.yexmp.2015.03.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 03/09/2015] [Indexed: 10/23/2022]
Abstract
We have previously shown that RGC-32 is involved in cell cycle regulation in vitro. To define the in vivo role of RGC-32, we generated RGC-32 knockout mice. These mice developed normally and did not spontaneously develop overt tumors. To assess the effect of RGC-32 deficiency on cell cycle activation in T cells, we determined the proliferative rates of CD4(+) and CD8(+) T cells from the spleens of RGC-32(-/-) mice, as compared to wild-type (WT, RGC-32(+/+)) control mice. After stimulation with anti-CD3/anti-CD28, CD4(+) T cells from RGC-32(-/-) mice displayed a significant increase in [(3)H]-thymidine incorporation when compared to WT mice. In addition, both CD4(+) and CD8(+) T cells from RGC-32(-/-) mice displayed a significant increase in the proportion of proliferating Ki67(+) cells, indicating that in T cells, RGC-32 has an inhibitory effect on cell cycle activation induced by T-cell receptor/CD28 engagement. Furthermore, Akt and FOXO1 phosphorylation induced in stimulated CD4(+) T-cells from RGC-32(-/-) mice were significantly higher, indicating that RGC-32 inhibits cell cycle activation by suppressing FOXO1 activation. We also found that IL-2 mRNA and protein expression were significantly increased in RGC-32(-/-) CD4(+) T cells when compared to RGC-32(+/+) CD4(+) T cells. In addition, the effect of RGC-32 on the cell cycle and IL-2 expression was inhibited by pretreatment of the samples with LY294002, indicating a role for phosphatidylinositol 3-kinase (PI3K). Thus, RGC-32 is involved in controlling the cell cycle of T cells in vivo, and this effect is mediated by IL-2 in a PI3K-dependent fashion.
Collapse
Affiliation(s)
- Cosmin A Tegla
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA; Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, USA
| | - Cornelia D Cudrici
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Jacob Danoff
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Adam M Kruszewski
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Armugam P Mekala
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Sonia I Vlaicu
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA; Department of Internal Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ching Chen
- Department of Pathology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Tudor C Badea
- Retinal Circuit Development and Genetics Unit, N-NRL, National Eye Institute, Bethesda, MD, USA
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, USA; Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, USA; Veterans Administration Multiple Sclerosis Center of Excellence, Baltimore, MD, USA.
| |
Collapse
|
23
|
Ai H, Yang B, Li J, Xie X, Chen H, Ren J. Population history and genomic signatures for high-altitude adaptation in Tibetan pigs. BMC Genomics 2014; 15:834. [PMID: 25270331 PMCID: PMC4197311 DOI: 10.1186/1471-2164-15-834] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 09/25/2014] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The Tibetan pig is one of domestic animals indigenous to the Qinghai-Tibet Plateau. Several geographically isolated pig populations are distributed throughout the Plateau. It remained an open question if these populations have experienced different demographic histories and have evolved independent adaptive loci for the harsh environment of the Plateau. To address these questions, we herein investigated ~ 40,000 genetic variants across the pig genome in a broad panel of 678 individuals from 5 Tibetan geographic populations and 34 lowland breeds. RESULTS Using a series of population genetic analyses, we show that Tibetan pig populations have marked genetic differentiations. Tibetan pigs appear to be 3 independent populations corresponding to the Tibetan, Gansu and Sichuan & Yunnan locations. Each population is more genetically similar to its geographic neighbors than to any of the other Tibetan populations. By applying a locus-specific branch length test, we identified both population-specific and -shared candidate genes under selection in Tibetan pigs. These genes, such as PLA2G12A, RGCC, C9ORF3, GRIN2B, GRID1 and EPAS1, are involved in high-altitude physiology including angiogenesis, pulmonary hypertension, oxygen intake, defense response and erythropoiesis. A majority of these genes have not been implicated in previous studies of highlanders and high-altitude animals. CONCLUSION Tibetan pig populations have experienced substantial genetic differentiation. Historically, Tibetan pigs likely had admixture with neighboring lowland breeds. During the long history of colonization in the Plateau, Tibetan pigs have developed a complex biological adaptation mechanism that could be different from that of Tibetans and other animals. Different Tibetan pig populations appear to have both distinct and convergent adaptive loci for the harsh environment of the Plateau.
Collapse
Affiliation(s)
- Huashui Ai
- Key Laboratory for Animal Biotechnology of Jiangxi Province and the Ministry of Agriculture of China, Jiangxi Agricultural University, Nanchang, 330045 P. R China
| | - Bin Yang
- Key Laboratory for Animal Biotechnology of Jiangxi Province and the Ministry of Agriculture of China, Jiangxi Agricultural University, Nanchang, 330045 P. R China
| | - Jing Li
- Key Laboratory for Animal Biotechnology of Jiangxi Province and the Ministry of Agriculture of China, Jiangxi Agricultural University, Nanchang, 330045 P. R China
| | - Xianhua Xie
- Key Laboratory for Animal Biotechnology of Jiangxi Province and the Ministry of Agriculture of China, Jiangxi Agricultural University, Nanchang, 330045 P. R China
| | - Hao Chen
- Key Laboratory for Animal Biotechnology of Jiangxi Province and the Ministry of Agriculture of China, Jiangxi Agricultural University, Nanchang, 330045 P. R China
| | - Jun Ren
- Key Laboratory for Animal Biotechnology of Jiangxi Province and the Ministry of Agriculture of China, Jiangxi Agricultural University, Nanchang, 330045 P. R China
| |
Collapse
|
24
|
Guo S, Philbrick MJ, An X, Xu M, Wu J. Response gene to complement 32 (RGC-32) in endothelial cells is induced by glucose and helpful to maintain glucose homeostasis. Int J Clin Exp Med 2014; 7:2541-2549. [PMID: 25356107 PMCID: PMC4211757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/26/2014] [Indexed: 06/04/2023]
Abstract
Endothelium dysfunction has been understood primarily in terms of abnormal vasomotor function, which plays an important role in the pathogenesis of diabetes and chronic diabetic complications. However, it has not been fully studied that the endothelium may regulate metabolism itself. The response gene to complement 32 (RGC-32) has be considered as an angiogenic inhibitor in the context of endothelial cells. We found that RGC-32 was induced by high fat diet in vivo and by glucose or insulin in endothelial cells, and then we set out to investigate the role of endothelial RGC-32 in metabolism. DNA array analysis and qPCR results showed that glutamine-fructose-6-phosphate aminotransferase [isomerizing] 1 (GFPT1), solute carrier family 2 (facilitated glucose transporter), member 12 (SLC2A12, GLUT12) and glucagon-like peptide 2 receptor (GLP2R) may be among possible glucose metabolism related downstream genes of RGC-32. Additionally, in the mice with endothelial specific over-expressed RGC-32, the disposal of carbohydrate was improved without changing insulin sensitivity when mice were faced with high fat diet challenges. Taken together, our findings suggest that RGC-32 in the endothelial cells regulates glucose metabolism related genes and subsequent helps to maintain the homeostasis of blood glucose.
Collapse
Affiliation(s)
- Shuzhen Guo
- School of Preclinical Medicine, Beijing University of Chinese MedicineBeijing, China
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| | - Melissa J Philbrick
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| | - Xiaojing An
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| | - Ming Xu
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| | - Jiaping Wu
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBoston, MA, USA
| |
Collapse
|
25
|
Vlaicu SI, Tegla CA, Cudrici CD, Danoff J, Madani H, Sugarman A, Niculescu F, Mircea PA, Rus V, Rus H. Role of C5b-9 complement complex and response gene to complement-32 (RGC-32) in cancer. Immunol Res 2013; 56:109-21. [PMID: 23247987 DOI: 10.1007/s12026-012-8381-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Complement system activation plays an important role in both innate and acquired immunity, with the activation of complement and the subsequent formation of C5b-9 terminal complement complex on cell membranes inducing target cell death. Recognition of this role for C5b-9 leads to the assumption that C5b-9 might play an antitumor role. However, sublytic C5b-9 induces cell cycle progression by activating signal transduction pathways and transcription factors in cancer cells, indicating a role in tumor promotion for this complement complex. The induction of the cell cycle by C5b-9 is dependent upon the activation of the phosphatidylinositol 3-kinase (PI3K)/Akt/FOXO1 and ERK1 pathways in a Gi protein-dependent manner. C5b-9 also induces response gene to complement (RGC)-32, a gene that plays a role in cell cycle promotion through activation of Akt and the CDC2 kinase. RGC-32 is expressed by tumor cells and plays a dual role in cancers, in that it has both a tumor suppressor role and tumor-promoting activity. Thus, through the activation of tumor cells, the C5b-9-mediated induction of the cell cycle plays an important role in tumor proliferation and oncogenesis.
Collapse
Affiliation(s)
- Sonia I Vlaicu
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Cui XB, Guo X, Chen SY. Response gene to complement 32 deficiency causes impaired placental angiogenesis in mice. Cardiovasc Res 2013; 99:632-9. [PMID: 23695833 DOI: 10.1093/cvr/cvt121] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
AIMS The objectives of this study are to determine the role of response gene to complement 32 (RGC-32) in the placental angiogenesis during pregnancy and explore the underlying mechanisms. METHODS AND RESULTS RGC-32-deficient (RGC32(-/-)) mice were generated from C57BL/6 embryonic stem cells with deletion of exon 2 and 3 of the RGC-32 gene. Most of the RGC32(-/-) mice can survive. However, their body sizes were much smaller compared with their wild-type littermates when they were born. By examining the embryo development and placentas at 16.5 days post-coitum, we found that RGC32(-/-) embryos and foetal placentas were significantly smaller than the wild-type. Further analysis showed that the labyrinth zone of RGC32(-/-) placenta was smaller with defective angiogenesis. Mechanistically, vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) and placental growth factor (PlGF) were significantly down-regulated in RGC32(-/-) placentas, suggesting that VEGFR2 and PlGF may mediate RGC-32 function in placental angiogenesis. Indeed, knockdown of RGC-32 by shRNA inhibited VEGF-induced endothelial cell proliferation, migration, and tube formation while blocking VEGFR2 expression. RGC-32 appeared to regulate VEGFR2 expression via activation of NF-kB. Moreover, RGC-32 regulated trophoblasts proliferation via control of PlGF expression. CONCLUSION Absence of RGC-32 caused foetal growth restriction through interrupting the placental angiogenesis, which was due to the decrease in VEGFR2 expression through the NF-kB-dependent pathway in endothelial cells and PlGF expression in trophoblasts.
Collapse
Affiliation(s)
- Xiao-Bing Cui
- Department of Physiology and Pharmacology, University of Georgia, Athens, 30602, USA
| | | | | |
Collapse
|
27
|
Liu D, Liu X, Xing M. Epigenetic genes regulated by the BRAFV600E signaling are associated with alterations in the methylation and expression of tumor suppressor genes and patient survival in melanoma. Biochem Biophys Res Commun 2012; 425:45-50. [PMID: 22820187 DOI: 10.1016/j.bbrc.2012.07.046] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 07/11/2012] [Indexed: 01/20/2023]
Abstract
We have previously reported that the BRAFV600E signaling causes genome-wide aberrations in gene methylation in melanoma cells. To explore the potential molecular mechanisms for this epigenetic effect of BRAFV600E, in this in silico study we analyzed 11 microarray datasets retrieved from NCBI GEO database and examined the relationship of the expression of the epigenetic genes (genes involved in epigenetic regulation) with BRAFV600E signaling, methylation and expression of tumor-suppressor genes (TSGs) in melanoma, and patient survival with this cancer. Among 273 epigenetic genes examined, 12 genes were down-regulated (named DD genes) and 16 were up-regulated (UU genes) by suppression of the BRAFV600E signaling using inhibitors. While the expression of 245 non-DD/UU genes overall had no correlation with the expression and methylation of a set of potential TSGs, the expression of DD genes was significantly correlated negatively with the TSG expression and positively with TSG methylation. Expression of UU genes was positively, albeit weakly, associated with the TSG expression. Overall, no correlation was found between UU gene expression and TSG methylation. Importantly, the expression of DD genes, but not UU genes, was significantly associated with decreased survival of patients with melanoma. Interestingly, the promoters of DD genes contain more binding motifs of c-fos and myc, two BRAFV600E signaling-related transcription factors, than those of UU and non-DD/UU genes. Thus, these results link epigenetic genes to methylation and suppression of tumor suppressor genes as a mechanism involved in BRAFV600E-promoted melanoma tumorigenesis and uncover a novel molecular signature that predicts a poor prognosis of melanoma.
Collapse
Affiliation(s)
- Dingxie Liu
- Division of Endocrinology and Metabolism, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | | | | |
Collapse
|
28
|
An X, Jin Y, Philbrick MJ, Wu J, Messmer-Blust A, Song X, Cully BL, He P, Xu M, Duffy HS, Li J. Endothelial cells require related transcription enhancer factor-1 for cell-cell connections through the induction of gap junction proteins. Arterioscler Thromb Vasc Biol 2012; 32:1951-9. [PMID: 22652601 DOI: 10.1161/atvbaha.112.250159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Capillary network formation represents a specialized endothelial cell function and is a prerequisite to establish a continuous vessel lumen. Formation of endothelial cell connections that form the vascular structure is regulated, at least in part, at the transcriptional level. We report here that related transcription enhancer factor-1 (RTEF-1) plays an important role in vascular structure formation. METHODS AND RESULTS Knockdown of RTEF-1 by small interfering RNA or blockage of RTEF-1 function by the transcription enhancer activators domain decreased endothelial connections in a Matrigel assay, whereas overexpression of RTEF-1 in endothelial cells resulted in a significant increase in cell connections and aggregation. In a model of oxygen-induced retinopathy, endothelial-specific RTEF-1 overexpressing mice had enhanced angiogenic sprouting and vascular structure remodeling, resulting in the formation of a denser and more highly interconnected superficial capillary plexus. Mechanistic studies revealed that RTEF-1 induced the expression of functional gap junction proteins including connexin 43, connexin 40, and connexin 37. Blocking connexin 43 function inhibited RTEF-1-induced endothelial cell connections and aggregation. CONCLUSIONS These findings provide novel insights into the transcriptional control of endothelial function in the coordination of cell-cell connections.
Collapse
Affiliation(s)
- Xiaojin An
- Institute of Molecular Medicine, Peking University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lu J, Mitra S, Wang X, Khaidakov M, Mehta JL. Oxidative stress and lectin-like ox-LDL-receptor LOX-1 in atherogenesis and tumorigenesis. Antioxid Redox Signal 2011; 15:2301-33. [PMID: 21338316 DOI: 10.1089/ars.2010.3792] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) has been identified as a major receptor for oxidized low-density lipoprotein (ox-LDL) in endothelial cells, monocytes, platelets, cardiomyocytes, and vascular smooth muscle cells. Its expression is minimal under physiological conditions but can be induced under pathological conditions. The upregulation of LOX-1 by ox-LDL appears to be important for physiologic processes, such as endothelial cell proliferation, apoptosis, and endothelium remodeling. Pathophysiologic effects of ox-LDL in atherogenesis have also been firmly established, including endothelial cell dysfunction, smooth muscle cell growth and migration, monocyte transformation into macrophages, and finally platelet aggregation-seen in atherogenesis. Recent studies show a positive correlation between increased serum ox-LDL levels and an increased risk of colon, breast, and ovarian cancer. As in atherosclerosis, ox-LDL and its receptor LOX-1 activate the inflammatory pathway through nuclear factor-kappa B, leading to cell transformation. LOX-1 is important for maintaining the transformed state in developmentally diverse cancer cell lines and for tumor growth, suggesting a molecular connection between atherogenesis and tumorigenesis.
Collapse
Affiliation(s)
- Jingjun Lu
- Cardiovascular Division, VA Medical Center, University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
| | | | | | | | | |
Collapse
|
30
|
Zhang C, Lu L, Li Y, Wang X, Zhou J, Liu Y, Fu P, Gallicchio MA, Bach LA, Duan C. IGF binding protein-6 expression in vascular endothelial cells is induced by hypoxia and plays a negative role in tumor angiogenesis. Int J Cancer 2011; 130:2003-12. [PMID: 21618524 DOI: 10.1002/ijc.26201] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 05/06/2011] [Indexed: 12/12/2022]
Abstract
Hypoxia stimulates tumor angiogenesis by inducing the expression of angiogenic molecules. The negative regulators of this process, however, are not well understood. Here, we report that hypoxia induced the expression of insulin-like growth factor binding protein-6 (IGFBP-6), a tumor repressor, in human and rodent vascular endothelial cells (VECs) via a hypoxia-inducible factor (HIF)-mediated mechanism. Addition of human IGFBP-6 to cultured human VECs inhibited angiogenesis in vitro. An IGFBP-6 mutant with at least 10,000-fold lower binding affinity for IGFs was an equally potent inhibitor of angiogenesis, suggesting that this action of IGFBP-6 is IGF-independent. The functional relationship between IGFBP-6 and vascular endothelial growth factor (VEGF), a major hypoxia-inducible angiogenic molecule, was examined. While VEGF alone increased angiogenesis in vitro, co-incubation with IGFBP-6 abolished VEGF-stimulated angiogenesis. The in vivo role of IGFBP-6 in angiogenesis was tested in flk1:GFP zebrafish embryos, which exhibit green fluorescence protein in developing vascular endothelium, permitting visualization of developing blood vessels. Injection of human IGFBP-6 mRNA reduced the number of embryonic inter-segmental blood vessels by ∼40%. This anti-angiogenic activity is conserved in zebrafish because expression of zebrafish IGFBP-6b had similar effects. To determine the anti-angiogenic effect of IGFBP-6 in a tumor model, human Rh30 rhabdomyosarcoma cells stably transfected with IGFBP-6 were inoculated into athymic BALB/c nude mice. Vessel density was 52% lower in IGFBP-6-transfected xenografts than in vector control xenografts. These results suggest that the expression of IGFBP-6 in VECs is up-regulated by hypoxia and IGFBP-6 inhibits angiogenesis in vitro and in vivo.
Collapse
Affiliation(s)
- Chunyang Zhang
- Key Laboratory of Marine Drugs (Ocean University of China), Ministry of Education and School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Huang WY, Xie W, Guo X, Li F, Jose PA, Chen SY. Smad2 and PEA3 cooperatively regulate transcription of response gene to complement 32 in TGF-β-induced smooth muscle cell differentiation of neural crest cells. Am J Physiol Cell Physiol 2011; 301:C499-506. [PMID: 21613609 PMCID: PMC3154553 DOI: 10.1152/ajpcell.00480.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 05/20/2011] [Indexed: 11/22/2022]
Abstract
Response gene to complement 32 (RGC-32) is activated by transforming growth factor- β (TGF-β) and plays an important role in smooth muscle cell (SMC) differentiation from neural crest Monc-1 cells. The molecular mechanism governing TGF-β activation of RGC-32, however, remains to be determined. The present studies indicate that TGF-β regulates RGC-32 gene transcription. Sequence analysis revealed a Smad binding element (SBE) located in the region from -1344 to -1337 bp upstream of the transcription start site of RGC-32 gene. A polyomavirus enhancer activator (PEA3) binding site is adjacent to the SBE. Mutation at either SBE or PEA3 site significantly inhibited RGC-32 promoter activity. Mutations at both sites completely abolished TGF-β-induced promoter activity. Biochemically, TGF-β stimulated recruitment of Smad2, Smad4, and PEA3 to the RGC-32 promoter, as revealed by gel shift and chromatin immunoprecipitation analyses. Functionally, Smad2, but not Smad3, activated RGC-32 promoter. PEA3 appeared to enhance Smad2 activity. In agreement with their function, Smad2, but not Smad3, physically interacted with PEA3. In TGF-β-induced SMC differentiation of Monc-1 cells, knockdown of Smad2 by short hairpin RNA resulted in downregulation of RGC-32 and SMC marker genes. The downregulation of SMC markers, however, was rescued by exogenously introduced RGC-32. These results demonstrate that Smad2 regulation of RGC-32 transcription is essential for SMC differentiation from neural crest cells.
Collapse
Affiliation(s)
- Wen-Yan Huang
- Department of Physiology and Pharmacology, The University of Georgia, Athens, GA 30602, USA
| | | | | | | | | | | |
Collapse
|
32
|
Wang JN, Shi N, Xie WB, Guo X, Chen SY. Response gene to complement 32 promotes vascular lesion formation through stimulation of smooth muscle cell proliferation and migration. Arterioscler Thromb Vasc Biol 2011; 31:e19-26. [PMID: 21636805 DOI: 10.1161/atvbaha.111.230706] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVE The objectives of this study were to determine the role of response gene to complement 32 (RGC-32) in vascular lesion formation after experimental angioplasty and to explore the underlying mechanisms. METHODS AND RESULTS Using a rat carotid artery balloon-injury model, we documented for the first time that neointima formation was closely associated with a significantly increased expression of RGC-32 protein. Short hairpin RNA knockdown of RGC-32 via adenovirus-mediated gene delivery dramatically inhibited the lesion formation by 62% as compared with control groups 14 days after injury. Conversely, RGC-32 overexpression significantly promoted the neointima formation by 33%. Gain- and loss-of-function studies in primary culture of rat aortic smooth muscle cells (RASMCs) indicated that RGC-32 is essential for both the proliferation and migration of RASMCs. RGC-32 induced RASMC proliferation by enhancing p34(CDC2) activity. RGC-32 stimulated the migration of RASMC by inducing focal adhesion contact and stress fiber formation. These effects were caused by the enhanced rho kinase II-α activity due to RGC-32-induced downregulation of Rad GTPase. CONCLUSIONS RGC-32 plays an important role in vascular lesion formation following vascular injury. Increased RGC-32 expression in vascular injury appears to be a novel mechanism underlying the migration and proliferation of vascular smooth muscle cells. Therefore, targeting RGC-32 is a potential therapeutic strategy for the prevention of vascular remodeling in proliferative vascular diseases.
Collapse
Affiliation(s)
- Jia-Ning Wang
- Department of Physiology and Pharmacology, University of Georgia, Athens, 30602, USA
| | | | | | | | | |
Collapse
|
33
|
Suzuki H, Tanabe H, Mizukami H, Inoue M. Differential gene expression in rat vascular smooth muscle cells following treatment with coptisine exerts a selective antiproliferative effect. JOURNAL OF NATURAL PRODUCTS 2011; 74:634-638. [PMID: 21401114 DOI: 10.1021/np100645d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
It is known that coptisine (1), an isoquinoline alkaloid, selectively inhibits proliferation of rat primary vascular smooth muscle cells (VSMCs). In the present study, the characteristics of its antiproliferative effect on several types of smooth muscle-like cells were investigated and compared to the effects of berberine (2) and palmatine (3). To clarify further the mechanism underlying the VSMC-selective antiproliferative effect of 1, the genes responsible were investigated by determining which mRNAs showed expression regulated by 1. Coptisine (1) showed a greater antiproliferative effect on smooth muscle cells derived from the aorta than on those derived from other organs. Analysis of the mRNA expression revealed that 1 upregulated two genes, growth arrest and DNA-damage-inducible alpha (Gadd45a) and response gene to complement32 (Rgc32). Both genes remained unchanged in 3Y1 fibroblasts and were not affected by 2 and 3. Coptisine (1) was found to induce the mRNA of the Gadd45a and Rgc32 genes, specifically in VSMC. Activation of these genes by 1 may mediate inhibition of cell-cycle progression. However, as these genes are commonly expressed in various cell types, a selective target for 1 activity is likely to exist upstream of these genes.
Collapse
Affiliation(s)
- Hiroka Suzuki
- Laboratory of Medicinal Resources, School of Pharmacy, Aichi Gakuin University, 1-100 Kusumoto-cho, Chikusa-ku, Nagoya 464-8650, Japan
| | | | | | | |
Collapse
|
34
|
Guo X, Jose PA, Chen SY. Response gene to complement 32 interacts with Smad3 to promote epithelial-mesenchymal transition of human renal tubular cells. Am J Physiol Cell Physiol 2011; 300:C1415-21. [PMID: 21307346 DOI: 10.1152/ajpcell.00204.2010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previous studies demonstrate that response gene to complement 32 (RGC-32) mediates transforming growth factor-β(1)-induced epithelial-mesenchymal transition (EMT) of human renal proximal tubular cells. However, the mechanisms underlying RGC-32 function remain largely unknown. In the present study, we found that RGC-32 function in EMT is associated with Smad3. Coexpression of RGC-32 and Smad3, but not Smad2, induces a higher mesenchymal marker α-smooth muscle actin (α-SMA) protein expression as compared with RGC-32 or Smad3 alone, while knockdown of Smad3 using short hairpin interfering RNA blocks RGC-32-induced α-SMA expression. These data suggest that RGC-32 interacts with Smad3, but not Smad2, in the regulation of EMT. In addition to α-SMA, RGC-32 and Smad3 also synergistically activate the expression of extracellular matrix protein fibronectin and downregulate the epithelial marker E-cadherin. RGC-32 colocalizes with Smad3 in the nuclei of renal proximal tubular cells. Coimmunoprecipitation assays showed that Smad3, but not Smad2, physically interacts with RGC-32 in renal proximal tubular cells. Mechanistically, RGC-32 and Smad3 coordinate the induction of EMT by regulating the EMT regulators Slug and Snail. Taken together, our data demonstrate for the first time that RGC-32 interacts with Smad3 to mediate the EMT of human renal proximal tubular cells.
Collapse
Affiliation(s)
- Xia Guo
- Dept. of Physiology & Pharmacology, The University of Georgia, 501 D.W. Brooks Drive, Athens, GA 30602, USA
| | | | | |
Collapse
|
35
|
Abstract
The regulation of angiogenesis by hypoxia is an essential homeostatic mechanism that depends on a precise balance between positive and negative angiogenic regulatory molecules. Proangiogenic factors are well characterized; however, several in vivo and in vitro studies indicate that there are feedback mechanisms in place to inhibit angiogenesis during hypoxia. Understanding the signaling pathways leading to the negative feedback of angiogenesis will undoubtedly provide important tools to develop novel therapeutic strategies not only to enhance the angiogenic response in coronary artery disease but also to hinder deregulated angiogenesis in tumorigenesis.
Collapse
Affiliation(s)
- Angela Messmer-Blust
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | |
Collapse
|