4000-520-616
欢迎来到免疫在线!(蚂蚁淘生物旗下平台)  请登录 |  免费注册 |  询价篮
主营:主营:蛋白质免疫试剂
咨询热线电话
4000-520-616
当前位置: 首页 > 新闻动态 >
新闻详情
CIP, a Cardiac Isl1-Interacting Protein, Represses...
来自 : 发布时间:2024-05-09
This site uses cookies. By continuing to browse this site you are agreeing to our use of cookies.Click here for more information. ShareShare onFacebookTwitterLinked InMendeleyRedditDiggEmailJump toSupplementary MaterialsFree AccessResearch ArticlePDF/EPUBCIP, a Cardiac Isl1-Interacting Protein, Represses Cardiomyocyte Hypertrophy Zhan-Peng Huang, Hee Young Seok, Bin Zhou, Jinghai Chen, Jian-Fu Chen, Yazhong Tao, William T. Pu, and Da-Zhi Wang Zhan-Peng HuangZhan-Peng Huang From the Department of Cardiology (Z.P.H., H.Y., B.Z., J.C., W.T.P., D.Z.W.), Children\'s Hospital Boston, Harvard Medical School, Boston, Massachusetts; Department of Cell and Developmental Biology (Y.T., J.F.C., D.Z.W.), McAllister Heart Institute (Y.T., D.Z.W.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina; Key Laboratory of Nutrition and Metabolism (B.Z.), Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.Search for more papers by this author, Hee Young SeokHee Young Seok From the Department of Cardiology (Z.P.H., H.Y., B.Z., J.C., W.T.P., D.Z.W.), Children\'s Hospital Boston, Harvard Medical School, Boston, Massachusetts; Department of Cell and Developmental Biology (Y.T., J.F.C., D.Z.W.), McAllister Heart Institute (Y.T., D.Z.W.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina; Key Laboratory of Nutrition and Metabolism (B.Z.), Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.Search for more papers by this author, Bin ZhouBin Zhou From the Department of Cardiology (Z.P.H., H.Y., B.Z., J.C., W.T.P., D.Z.W.), Children\'s Hospital Boston, Harvard Medical School, Boston, Massachusetts; Department of Cell and Developmental Biology (Y.T., J.F.C., D.Z.W.), McAllister Heart Institute (Y.T., D.Z.W.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina; Key Laboratory of Nutrition and Metabolism (B.Z.), Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.Search for more papers by this author, Jinghai ChenJinghai Chen From the Department of Cardiology (Z.P.H., H.Y., B.Z., J.C., W.T.P., D.Z.W.), Children\'s Hospital Boston, Harvard Medical School, Boston, Massachusetts; Department of Cell and Developmental Biology (Y.T., J.F.C., D.Z.W.), McAllister Heart Institute (Y.T., D.Z.W.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina; Key Laboratory of Nutrition and Metabolism (B.Z.), Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.Search for more papers by this author, Jian-Fu ChenJian-Fu Chen From the Department of Cardiology (Z.P.H., H.Y., B.Z., J.C., W.T.P., D.Z.W.), Children\'s Hospital Boston, Harvard Medical School, Boston, Massachusetts; Department of Cell and Developmental Biology (Y.T., J.F.C., D.Z.W.), McAllister Heart Institute (Y.T., D.Z.W.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina; Key Laboratory of Nutrition and Metabolism (B.Z.), Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.Search for more papers by this author, Yazhong TaoYazhong Tao From the Department of Cardiology (Z.P.H., H.Y., B.Z., J.C., W.T.P., D.Z.W.), Children\'s Hospital Boston, Harvard Medical School, Boston, Massachusetts; Department of Cell and Developmental Biology (Y.T., J.F.C., D.Z.W.), McAllister Heart Institute (Y.T., D.Z.W.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina; Key Laboratory of Nutrition and Metabolism (B.Z.), Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.Search for more papers by this author, William T. PuWilliam T. Pu From the Department of Cardiology (Z.P.H., H.Y., B.Z., J.C., W.T.P., D.Z.W.), Children\'s Hospital Boston, Harvard Medical School, Boston, Massachusetts; Department of Cell and Developmental Biology (Y.T., J.F.C., D.Z.W.), McAllister Heart Institute (Y.T., D.Z.W.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina; Key Laboratory of Nutrition and Metabolism (B.Z.), Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.Search for more papers by this author, and Da-Zhi WangDa-Zhi Wang From the Department of Cardiology (Z.P.H., H.Y., B.Z., J.C., W.T.P., D.Z.W.), Children\'s Hospital Boston, Harvard Medical School, Boston, Massachusetts; Department of Cell and Developmental Biology (Y.T., J.F.C., D.Z.W.), McAllister Heart Institute (Y.T., D.Z.W.), School of Medicine, University of North Carolina, Chapel Hill, North Carolina; Key Laboratory of Nutrition and Metabolism (B.Z.), Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.Search for more papers by this authorOriginally published16 Feb 2012https://doi.org/10.1161/CIRCRESAHA.111.259663Circulation Research. 2012;110:818–830Other version(s) of this articleYou are viewing the most recent version of this article. Previous versions: January 1, 2012: Previous Version 1AbstractRationale:Mammalian heart has minimal regenerative capacity. In response to mechanical or pathological stress, the heart undergoes cardiac remodeling. Pressure and volume overload in the heart cause increased size (hypertrophic growth) of cardiomyocytes. Whereas the regulatory pathways that activate cardiac hypertrophy have been well-established, the molecular events that inhibit or repress cardiac hypertrophy are less known.Objective:To identify and investigate novel regulators that modulate cardiac hypertrophy.Methods and Results:Here, we report the identification, characterization, and functional examination of a novel cardiac Isl1-interacting protein (CIP). CIP was identified from a bioinformatic search for novel cardiac-expressed genes in mouse embryonic hearts. CIP encodes a nuclear protein without recognizable motifs. Northern blotting, in situ hybridization, and reporter gene tracing demonstrated that CIP is highly expressed in cardiomyocytes of developing and adult hearts. Yeast two-hybrid screening identified Isl1, a LIM/homeodomain transcription factor essential for the specification of cardiac progenitor cells in the second heart field, as a cofactor of CIP. CIP directly interacted with Isl1, and we mapped the domains of these two proteins, which mediate their interaction. We show that CIP represses the transcriptional activity of Isl1 in the activation of the myocyte enhancer factor 2C. The expression of CIP was dramatically reduced in hypertrophic cardiomyocytes. Most importantly, overexpression of CIP repressed agonist-induced cardiomyocyte hypertrophy.Conclusions:Our studies therefore identify CIP as a novel regulator of cardiac hypertrophy.IntroductionComplex genetic pathways intimately regulate heart development and function.1 Much of our current understanding of how cardiac gene expression is controlled is at the level of transcriptional regulation, in which multiple tissue-specific transcription factors have been implicated in the control of gene expression during cardiomyocyte differentiation.2 Those include Nkx2.5, a homeobox protein,3–5 MEF2C, a member of the myocyte enhance factor-2 (MEF2) family of MADS-box transcription factors,6–11 and GATA4, a GATA family zinc finger protein.12,13 Some of the well-characterized cardiac genes regulated by these cardiac transcription factors include cardiac-specific atrial natriuretic factor and cardiac α-actin.13,14Two distinct sources of cardiac precursor cells from the \"primary” and \"secondary heart fields” participate in heart formation.15–17 Whereas the primary heart field is essential for the formation of the initial heart tube, additional cardiac precursor cells recruited from the secondary heart field contribute to the future right ventricle and outflow tract.18,19 Loss-of-function studies have defined genes whose activity is required in secondary heart field progenitors for morphogenesis of outflow tract and right ventricle.18,20 Among the genes with key function in secondary heart field is Isl1, a LIM/homeodomain transcription factor.21–24One of the major responses of the heart to biomechanical stress and pathological stimuli is to undergo cardiac hypertrophy, an increase in the thickness of the cardiac ventricular wall. At a cellular level, cardiac hypertrophy is defined as an increase of cardiomyocyte size.25,26 Initially, cardiac hypertrophy is an adaptive response that maintains cardiac output in the face of increased workload. However, chronic activation of hypertrophic pathways is associated with adverse consequences that may lead to heart failure and sudden death.27Cardiac hypertrophy occurs in response to a variety of mechanical, hemodynamic, hormonal, and pathological stimuli.28–30 Numerous studies have demonstrated that many signaling pathways and transcriptional networks that normally regulate different aspects of cell proliferation, differentiation, and survival are also involved in the induction of cardiac hypertrophy.1,29,31 Hypertrophic growth involves control of cardiomyocyte gene expression at multiple molecular levels. Key regulators of gene expression in cardiac myocytes such as members of the MEF2 and GATA families are involved in the control of gene expression during cardiomyocyte hypertrophy. Epigenetic regulation of gene expression, including histone modification by histone acetyltransferases and histone deacetylases to remodel chromatin, has been proven as another mean of hypertrophic regulation.32,33 Cardiac hypertrophy is also accompanied by reactivation of a set of cardiac fetal genes, including those that encode atrial natriuretic factor, B-type natriuretic peptide, and β myosin heavy chain.26 Reactivation of these fetal genes suggests that molecular pathways that control heart development are redeployed to regulate hypertrophic growth.34 The pathophysiology of cardiac hypertrophy has been extensively studied for decades and much is known about signaling pathways that activate cardiac hypertrophy.26,30 However, relatively less is known about how cardiac hypertrophy is repressed.We previously identified myocardin as a SAP (SAF-A/B, Acinus, PIAS) domain transcriptional regulator.35,36 Myocardin is expressed in both cardiomyocytes and a subset of vascular and visceral smooth muscle cell types.35,36 Myocardin does not to bind DNA directly, but rather forms a stable ternary complex with serum response factor bound to DNA.37,38 As a transcriptional cofactor of serum response factor, myocardin potently transactivates CArG box-containing reporter genes.36,39,40 Genetic studies revealed that myocardin and myocardin-related transcription factors play critical roles in a variety of biological processes, including vascular smooth muscle development, aortic vessel patterning, mammary myoepithelium formation, and others.41–43 In this study, we report the identification and characterization of a novel cardiac-specific nuclear protein, cardiac Isl1-interacting protein (CIP). CIP interacts with the LIM/homeodomain transcription factor Isl1. We showed that the expression of CIP was repressed in hypertrophic hearts and, most importantly, CIP represses cardiomyocyte hypertrophy.MethodsAn expanded Methods section is provided in the Online Supplement.ResultsIdentification of CIP, a Novel Gene From Mouse HeartsWe searched expressed sequence tag databases for novel sequences present in cardiac cDNA libraries. Previously, this approach succeeded in identifying myocardin as a novel cardiac-specific and smooth muscle-specific transcription cofactor for serum response factor.36 An additional candidate expressed sequence tag sequence recovered in the search (GenBank Accession Number AA919489) contained a short 273 nucleotide tag that was derived from a mouse embryonic day 13 (embryonic day 13.5) heart cDNA library. Using this sequence as a probe to screen a mouse embryonic heart cDNA library, we identified a larger cDNA sequence of 1336 nucleotides encoding a putative protein of 309 amino acids, which we name CIP (cardiac Isl1-interesting protein; Figure 1A). Genbank Database search indicated that this cDNA clone is similar to RIKEN cDNA clone 2310046A06Rik and is a novel alternative slicing-produced isoform of a recently reported protein called MLIP (muscle-enriched A-type lamin-interacting protein).44 In addition to the putative lamin-A interaction domain in its N-terminus, CIP contains an AT-hook DNA binding domain in its C-terminus (aa 212–224), suggesting that CIP could function as a DNA-binding transcriptional regulator (Figure 1A, C; Online Figure I). Furthermore, the CIP protein contains a putative asparagine glycosylation site (aa 223–226), a poly serine stretch (aa 225–229), and a tyrosine phosphorylation site (aa 254–260; Figure 1A). Northern blot analysis using an adult mouse multiple tissue blot demonstrated that the expression of CIP is restricted to the heart. Two major transcripts, ≈1.4 K and ≈3.4 K nucleotides, respectively, were detected in the heart and likely represent products of alternative splicing (Figure 1B). The cDNA sequence we have cloned represents the ≈1.4 kb transcript, whereas the sequences of the ≈3.4 kb remain to be identified.Download figureDownload PowerPointFigure 1. Identification of the CIP gene.A, Nucleotide sequences and deduced amino acid sequences. Nucleotides for open reading frame (ORF) are shown in uppercase; nucleotides for both 5′ and 3′ untranslated regions (UTRs) are shown in lowercase. The corresponding amino acids (in single letter code) are also shown. Amino acids underlined indicate the putative AT-hook domain and poly-Ser domain. The predicted amino acid modifications are also indicated and their recognition motifs are shown in boxes. B, Northern blotting analysis of CIP expression using RNAs of different tissues from adult mice. C, Gene structure and alternative splicing of the mouse CIP gene. Empty boxes indicate ORF, gray boxes indicate UTR. The color boxes in alternative splicing variants indicate putative domains shown in (A). D, CIP is a nuclear protein. Immunochemistry detecting the nuclear localization of the FLAG-CIP fusion proteins in transfected COS and Hela cells.The Cip gene contains at least eight exons and spans over 245 kb on mouse chromosome 9. Cip is evolutionary, conserved from fish to human (Online Figure II). Sequencing of Cip reverse-transcriptase polymerase chain reaction products identified complex alternative splicing (Figure 1C). The functional significance of those isoforms is currently not clear.Next, we examined the subcellular location of the CIP protein. We transiently overexpressed Flag-CIP fusion proteins in COS7 and Hela cells. Immunochemistry assays revealed that the Flag-CIP fusion proteins are primarily located in the nuclei of transfected cells. We also observed a weaker distribution of the Flag-CIP proteins in the cytoplasm (Figure 1D).CIP Expression Is Restricted to Cardiomyocytes of Developing HeartsTo define the expression pattern of Cip during development, in situ hybridization was performed using an antisense probe to the 3′ untranslated region of the mouse Cip transcript. Whole-mount in situ hybridization first demonstrated that CIP expression is restricted to the heart of embryonic day 9.5 mouse embryos, with highest expression level detected in the ventricle (Figure 2A). In situ hybridization on tissue sections of staged mouse embryos revealed that Cip expression was first detected in the heart of embryonic day 8.5 mouse embryos (Figure 2B). Cip continued to be restricted to the heart from embryonic day 9.5 to embryonic day 15.5 (Figure 2C, D, E, F, G). An apparently positive signal was also detected in the truck of embryonic day 9.5 embryo, which might represent transient expression of CIP in presomitic mesoderm (Figure 2C). From embryonic day 11.5 to embryonic day 15.5, Cip expression appeared to be higher in the ventricles of embryonic hearts (Figure 2E, F, G). Myocardial Cip expression continued in the adult heart (Figure 2H).Download figureDownload PowerPointFigure 2. Expression of CIP gene in embryonic and adult mouse tissues.A, Whole-mount in situ hybridization detecting CIP transcript expression in embryonic day 9.5 mouse embryos. In situ hybridization detecting CIP transcript expression on sections of staged mouse embryos (B–G) and in an adult mouse heart (H). B, E, F, G, Transverse sections. C, D, H, Sagittal sections. h, heart; a, atrium; la, left atrium; lv, left ventricle; ra, right atrium; rv, right ventricle; v, ventricle.Because the heart consists of cardiomyocyte, cardiac fibroblast, smooth muscle cell, endothelial cell, and epicardial cell, we performed additional experiments to determine exactly in which cell type/lineage the CIP is expressed. First, we utilized a genetic approach in which the Rosa-mT-mG reporter line was used to trace the expression of CIP in different cell types. The Rosa-mT-mG mice possess loxP sites on both sides of a membrane-targeted tdTomato (mT) cassette and express strong red fluorescence in all tissues and cell types (Figure 3A, B). The presence of Cre will lead to the deletion of the mT cassette and the activation of the downstream membrane-targeted enhanced green fluorescent protein (EGFP) (mG) cassette (Figure 3A). The membrane-targeted EGFP can be utilized as a marker for fluorescence-activated cell sorting (Figure 3D). To label and sort out the cardiomyocytes, the Rosa-mT-mG mice were bred with the cTNT-Cre mice, in which the Cre recombinase is driven by the cardiomyocyte-specific cardiac troponin T promoter (Figure 3B). Immunochemistry confirmed the labeling of cardiomyocyte in Rosa-mT-mG/cTNT-Cre embryos (Figure 3C). Hearts were dissected out from embryonic day 10.5 Rosa-mT-mG/cTNT-Cre embryos and digested. EGFP-positive cardiomyocytes and EGFP-negative noncardiomyocytes were separated by fluorescence-activated cell sorting (Figure 3D). Quantitative reverse-transcriptase polymerase chain reaction detected high expression level of Myh6 (α-myosin heavy chain) and CIP in the EGFP-positive cell sample but not in EGFP-negative cell sample, whereas endothelial markers Flk1, platelet/endothelial cell adhesion molecule 1, and fibrotic tissue marker Postn were highly expressed in EGFP-negative cell sample but not in the EGFP-positive cell sample (Figure 3E, F, G; n=3). These data, together with the results of in situ hybridization, suggested that CIP is expressed in cardiomyocytes of developing hearts.Download figureDownload PowerPointFigure 3. Quantitative reverse-transcriptase polymerase chain reaction (RT-PCR) analyses of CIP expression in cardiomyocytes of mouse embryos.A, Schematic of the RosamTmG/+ reporter system. In the presence of Cre recombinase, which deletes the mT cassette, the expression of membranous tomato red (mT) turns into membranous green fluorescence protein (mG). B, When the RosamTmG/+ reporter line was crossed with a cardiomyocyte-specific Cre line (TNT-Cre), membranous green fluorescent protein (GFP) is detected in embryonic day 10.5 heart (h), whereas there is no green signal in the heart of control littermate. Bar=500 μm. C, Membranous GFP is detected in NKX2.5-positive and TNNT2-positive myocardium, but not the platelet/endothelial cell adhesion molecule 1 (PECAM1), positive endocardium. Bar=10 μm. D, Fluorescence-activated cell sorting (FACS) isolation of GFP-positive and GFP-negative cell populations from TNT-Cre and RosamTmG/+ hearts. Insert shows that less than 0.01% GFP-positive cells were found in the hearts of control littermates. E, GFP-positive population is highly enriched for cardiomyocyte-specific marker Myh6, documented by quantitative RT-PCR assays. F, GFP-positive population expresses very low levels of endothelial marker Flk1 or Pecam1, and fibrotic tissue marker Postn, indicating the specificity of cell sorting. G, CIP is highly expressed in the GFP-positive cardiomyocytes, but not in GFP-negative noncardiomyocytes. Expression in GFP-negative population is set as 1. *P 0.01. Three embryos were used (n=3) for analysis in each group.Next, we used a Cip gene trap mouse line in which a LacZ reporter gene was inserted into the Cip genomic locus (see Methods for details) to further map the expression of Cip in the heart (Online Figure III). LacZ reporter gene expression was restricted to the developing heart at embryonic day 10.5 and embryonic day 13.5, recapitulating endogenous expression pattern of Cip and highlighting the cardiac specific expression of this gene (Figure 4A, K). Immunohistochemistry analyses clearly demonstrated that the expression of Cip, as marked by β-galactosidase, overlapped with that of Nkx2-5, cardiac troponin T (TNNT2), and cardiac actin (ACTN2) in cardiomyocytes (Figure 4B, C, D, G, J, yellow arrow heads). Cip-expressing cells did not coexpress WT1, a marker of epicardium, or platelet/endothelial cell adhesion molecule, a marker of endothelial cells (Figure 4E, F, H, I, white arrows and arrow heads; n 3). Together these studies clearly demonstrate that Cip is specifically expressed in cardiomyocytes during embryogenesis. Interestingly, we detected CIP expression in the skeletal muscle of neonatal mice (Online Figure IV, middle). However, the expression of CIP in skeletal muscle appears to be transient, because we did not detect its expression in embryonic skeletal muscle (Online Figure IV, left) or in adult skeletal muscle tissue using the lacZ reporter (Online Figure IV, right) or by Northern blot analyses (Figure 1B).Download figureDownload PowerPointFigure 4. Characterization of CIP expression using a LacZ reporter gene in mouse embryos.A, Whole-mount β-galactosidase staining of mouse embryonic day 10.5 embryos harboring a LacZ cassette inserted in the CIP locus (CIPlacZ/+), or the control (CIP+/+). Positive β-galactosidase staining is only detected in the heart. B–J, Immunohistochemistry using indicated antibodies on tissue sections of embryonic day 10.5 CIPlacZ/+ embryo. Note that CIP-positive cells (marked by antibodies against the β-galactosidase and shown in red) overlay with Nkx2-5 (D, G), cardiac α-actin (ACTN2; J), and cardiac troponin T-positive (TNNT2; B, C) cardiomyocytes (yellow arrowheads), but not that of WT1-positive epicardium (whitearrows; E, F) or platelet/endothelial cell adhesion molecule (PECAM)-positive fibroblasts (whitearrowheads; H, I). DAPI (blue) staining marks the nucleus. K, Whole-mount β-galactosidase staining of mouse embryonic day 13.5 embryonic hearts harboring a LacZ cassette inserted in the CIP locus (CIPlacZ/+) or the control (CIP+/+). L–Q, Immunohistochemistry using indicated antibodies on tissue sections of embryonic day 13.5 CIPlacZ/+ embryo. Note that CIP-positive cells (marked by antibodies against the β-galactosidase) overlay with cardiac α-actin (ACTN2) and cardiac troponin T-positive (TNNT2) cardiomyocytes, not that of WT1-positive epicardium or PECAM-positive fibroblasts. DAPI staining marks the nucleus. B, E, H, bar=100 μm; C, D, F, G, I, J, bar=10 μm; L, O, bar=200 μm; M, N, P, Q, bar=40 μm. Data are presented as representative images derived from more than three embryos. A, atrium; LV, left ventricle; RV, right ventricle.CIP Interacts With Cardiac Transcriptional Factor Islet1To identify putative CIP-interacting proteins, we performed yeast two-hybrid screening. The bait consisted of full-length CIP protein fused with the yeast GAL4 DNA binding domain. The bait construct did not autonomously activate transcription in yeast. We screened a mouse embryonic day 10.5 embryonic cDNA prey library and identified more than 30 positive clones, one of which encoded Islet1 (Isl1), a LIM/homeodomain transcription factor essential for differentiation of second heart field cardiac progenitors.23,45To confirm the interaction of CIP and Isl1, we performed coimmunoprecipitation assays. HEK293T cells were transfected with expression plasmids for FLAG-CIP, Myc-Isl1, or both. FLAG immunoprecipitates of cells expressing both constructs contained Isl1 protein, as demonstrated by Western blotting with anti-Myc antibodies. As negative controls, Isl1 was not detected in FLAG immunoprecipitates of cells transfected with each construct alone (Figure 5A). To further confirm direct interaction between CIP and Isl1, we performed in vitro GST–fusion protein pull-down assays. Isl1 was 35S-labeled in a cell-free translation system, and the labeled Isl1 protein was then incubated with either GST-CIP fusion protein or GST protein alone (to serve as a negative control). As expected, GST-CIP fusion protein, but not GST alone, bound Isl1 (Figure 5B).Download figureDownload PowerPointFigure 5. CIP interacts with Isl1 and represses its transcription activity.A, Coimmunoprecipitation assays showing the interaction between Flag-tagged CIP and Myc-tagged Isl1. HEK-293T cells were cotransfected Flag-CIP, Myc-Isl1, or both. Anti-Flag antibody was used to immunoprecipitate (IP) cell extracts and anti-Myc antibody was used in Western blot (WB) to detect Myc-Isl1 protein in the complex; 10% cell lysate was used as input to demonstrate the expression of tagged proteins (bottomtwo panels). B, In vitro GST-fusion protein pull-down assays showing the direct interaction between CIP and Isl1. Isl1 protein was synthesized in vitro and labeled with S-35. GST-CIP protein, but not GST protein, pulled down S-35–labeled Isl1; 20% of labeled Isl1 protein was load as control (input). C, Map the interaction domains of the CIP protein that mediate the interaction between Isl1 and CIP using coimmunoprecipitation assays. HEK-293T cells were cotransfected Flag-Isl1, Myc-CIP full-length (1–309), or indicated truncated mutants, or both. Anti-Flag antibody was used to immunoprecipitate (IP) cell extracts and anti-Myc antibody was used in Western blot (WB) to detect Myc-CIP protein in the complex; 10% cell lysate was used as input to demonstrate the expression of tagged proteins (lower two panels). The interaction domains are summarized in the bottom diagram. D, Map the interaction domains of Isl1 proteins that mediate the interaction between Isl1 and CIP using coimmunoprecipitation assays. HEK-293T cells were cotransfected Myc-CIP, Flag-Isl1 full-length (1–349), or indicated truncated mutants, or both. Anti-Flag antibody was used to IP cell extracts and anti-Myc antibody was used in Western blot (WB) to detect Myc-CIP protein in the complex; 10% cell lysate was used as input to demonstrate the expression of tagged proteins (lower two panels). The interaction domains are summarized in the bottom diagram. E, Isl1 and CIP are coexpressed in cardiomyocytes of outflow tract of mouse embryos. Immunohistochemistry to demonstrate that Isl1 (green) and β-galactosidase-positive CIP-expressing (red) cells are colocated in the cardiomyocytes of outflow tract (OFT) of embryonic day 10.5 mouse embryos (arrowheads). DAPI staining indicates the nucleus. Bar=50 μm. A, atrium; LV, left ventricle. F, Enlargement of boxed area in (E) shows that Isl1 and CIP expression is overlapped in the OFT cardiomyocytes (white arrowheads). The expression of Isl1 (green), CIP (as marked by β-galactosidase-positive cells, red), DAPI (blue), and merged images were shown. Isl1 and CIP-positive cells were indicated by white arrowheads. Note that Isl1 is not expressed in the cardiomyocytes of atrium where CIP is expressed (yellowarrows). G, CIP is a transcriptional repressor. HEK293T cells were transiently transfected with expression vectors encoding the CIP fused to GAL4 (1–147) and the pL8G5-luciferase reporter, which contains binding sites for the GAL4 DNA binding domain. Luciferase activity is expressed as relative activity in which the control was assigned a value of 1. Data represent the mean±SD from at least three independent experiments in triplicate. *P 0.05. H, CIP represses the transcription activity of Isl1. MEF2C enhancer-luciferase reporter was cotransfected with indicated expression plasmids in HEK293T cells and luciferase activity was measured. Results were presented as relative luciferase activity in which the control was assigned a value of 1. Data represent the mean±SD from at least three independent experiments in triplicate. *P 0.05. I, Define the domains of the CIP protein that mediate the repression of Isl1 transcription activity. MEF2C enhancer-luciferase reporter was cotransfected with indicated expression plasmids (full-length Isl1, full-length or truncated CIP mutants) in HEK293T cells and luciferase activity was measured. Results were presented as relative luciferase activity in which the control was assigned a value of 1. Data represent the mean±SD from at least three independent experiments in triplicate. *P 0.05. J, CIP represses myocardin-mediated and serum response factor-mediated transactivation of the atrial natriuretic factor (ANF)-luciferase reporter. The ANF-luciferase reporter was cotransfected with indicated expression plasmids in HEK293T cells and luciferase activity was measured. Results were presented as relative luciferase activity in which the control was assigned a value of 1. Data represent the mean±SD from at least three independent experiments in triplicate. *P 0.05.Next, we attempted to map the domains of these two proteins that mediate their interaction. We made series of deletion mutants and tested their interaction using coimmunoprecipitation assays. As shown inFigure 5C, the interaction between CIP and Isl1 is readily detected in full-length (1–309) and both C-terminal and N-terminal truncated mutants (1–200 and 100–309). However, further C-terminal deletion (1–100) abolished the interaction, suggesting that regions between aa 100 and 200 are required for CIP to interact with Isl1 (Figure 5C). Similarly, we examined domains in the Isl1 protein that are needed for the interaction with CIP. We found that aa 1 to 133 fragment is sufficient to mediate the interaction, whereas the aa 79 to 349 region failed to interact (Figure 5D). These data indicate that the N-terminal region of the Isl1 protein is essential for its interaction with the CIP protein.Isl1 is expressed in the cardiac progenitors and marks the second heart field. It was previously reported that Isl1 is not expressed in the cardiomyocytes of adult hearts.23 We investigated whether CIP and Isl1 are coexpressed in mouse embryos. The expression of Isl1 was examined in Cip-lacZ reporter embryos. As shown inFigures 5E and 5F, both LacZ and Isl1 were expressed in cardiomyocytes located in the outflow tract of embryonic day 10.5 mouse embryos (Figure 5E, F, white arrowheads). However, Isl1 is not expressed in the cardiomyocytes of atrium or in the left ventricle, where CIP is expressed (Figure 5F, yellow arrows). Given that Isl1 is expressed in cardiac progenitor cells of the second heart field and newly formed cardiomyocytes in the outflow tracts,23 these results confirmed that CIP and Isl1 are coexpressed in the cardiac progenitor cells of embryonic hearts.CIP Is a Transcription Cofactor of Isl1 and Regulates Its Transcriptional ActivityBecause CIP was localized to the nucleus, we asked whether it possessed transcriptional activity. We fused the full-length CIP protein to the DNA-binding domain of yeast GAL4 protein. Interestingly, this fusion protein repressed the GAL4-dependent reporter when cotransfected into HEK293T cells (Figure 5G). Similar, we observed that GAL4-CIP repressed the reporter in Hela and COS7 cells (data not shown).Loss-of-function studies indicate that Isl1 is essential for cardiac development.23 Isl1 activates the expression of the Mef2c gene by directly binding to its anterior heart field enhancer.46 We tested whether CIP participates in regulation of the Mef2c anterior heart field enhancer. As expected, Isl1 constantly activated the Mef2c enhancer luciferase reporter, although its activity does not appear to be potent (Figure 5I). CIP alone did not significantly affect Mef2c enhancer activity. However, coexpression of Isl1 (the full-length, 1–309) and CIP inhibited Isl1-mediated Mef2c enhancer luciferase activity (Figure 5I). Further analyses, using different CIP deletion mutants, showed that the aa 1 to 200 and the aa 100 to 309 constructs (but not the aa 1–100) were able to repress Isl1-mediated activation of the Mef2c enhancer. These observations indicate that direct interaction of CIP and Isl1 is necessary for CIP to repress Isl1. Our results suggest that CIP is a transcription cofactor and represses the transcriptional activity of Isl1.Next, we tested whether CIP represses the activity of other transcriptional regulators. Myocardin is a transcription factor specifically expressed in cardiac and smooth muscle that potently stimulates atrial natriuretic factor promoter activity in conjunction with the transcription factor serum response factor.36 We found that CIP significantly repressed myocardin-mediated activation of the atrial natriuretic factor–luciferase reporter (Figure 5J). Similarly, CIP also repressed serum response factor-dependent activation of the atrial natriuretic factor–luciferase reporter (Figure 5J). Together, our results indicated that CIP is a putative transcriptional repressor expressed in the heart.The Expression of CIP Is Dysregulated in Hypertrophic HeartsThe heart undergoes cardiac remodeling in response to cardiac injury or stress. Increased pressure or volume overload in the heart results in cardiac hypertrophy, a process in which the size of cardiomyocytes increases without an increase in the number of cardiomyocytes. Interestingly, MEF2C was previously shown to be a key regulator of cardiac hypertrophy and dilated cardiomyopathy.11,47–50 Because CIP repressed the Mef2c enhancer activity, we hypothesized that CIP regulates cardiac hypertrophy. We first examined whether hypertrophic signals regulate cardiac CIP expression. We used a transverse aortic constriction to induce pressure overload and subsequent pathological cardiac hypertrophy. As expected, atrial natriuretic peptide and B-type natriuretic peptide, well-known hypertrophy markers, were dramatically upregulated in transverse aortic constriction mice at day 3 and day 7 (Figure 6A). In contrast, Cip transcripts were substantially reduced in transverse aortic constriction mice. Similarly, expression of CIP was reduced in a second well-characterized cardiac hypertrophy model,27 in which transgenic expression of activated calcineurin stimulates dramatic cardiac hypertrophy (Figure 6B).Download figureDownload PowerPointFigure 6. CIP represses cardiomyocyte hypertrophy.A, Gene expression of CIP and hypertrophic marker, atrial natriuretic peptide (ANP), and B-type natriuretic peptide (BNP), determined by quantitative polymerase chain reaction (qPCR) in heart samples from sham group and transverse aortic constriction (TAC) group at two different time point (3 days and 7 days). n=4. B, Gene expression of CIP and hypertrophic marker ANP determined by qPCR in heart samples from 1-month-old Myh6-calcineurin transgenic mice (CnA) and control littermates (wild-type [WT]). n=3. C, Western blotting analysis showing decreased expression of CIP proteins in neonatal rat cardiomyocytes, which were induced to develop hypertrophy by different hypertrophic agonists in vitro. No treated sample serves as a control. LIF, leukemia inhibitory factor; ET-1, endothelin-1; PE, phenylephrine. D, Quantitative reverse-transcriptase polymerase chain reaction (RT-PCR) showing CIP expression in rat neonatal cardiomyocytes with or without PE treatment. *P 0.05. E, CIP represses PE-induced hypertrophy in neonatal cardiomyocytes. Representative images of cardiomyocytes infected with adenoviral-CIP (ad-CIP) or adenoviral-green fluorescent protein (GFP) (ad-GFP), to serve as controls, and treated with PE (or without treatment to serve as controls). Anti-α-actinin antibodies were used to indicate cardiomyocytes (red). DAPI marks nucleus. Bar=21 μm. F, Quantitative analysis of cardiomyocyte cell size. Approximately 300 cells positive for α-actinin from each treatment were randomly chosen for surface area measurement. *P 0.05. G, Quantitative RT-PCR showing the downregulation of PE-induced expression of hypertrophic marker ANP, BNP, and β-myosin heavy chain (β-MHC) when CIP was overexpressed in neonatal cardiomyocytes. n=4. *P 0.05.In vivo cardiac hypertrophy models are complex and encompass both direct effects of inducing stimuli and secondary effects of myocardial responses, including cardiac fibrosis and failure. To directly examine the effect of hypertrophic stimuli on cardiomyocyte Cip expression, we used the well-established neonatal rat cardiomyocyte hypertrophy model. We treated neonatal rat cardiomyocytes with a panel of hypertrophic agonists and measured the effect on Cip expression. We found that the CIP protein level was significantly reduced in neonatal rat cardiomyocytes treated with the hypertrophic agonists leukemia inhibitory factor, endothelin-1, and phenylephrin (PE) (Figure 6C). Quantitative reverse-transcriptase polymerase chain reaction analyses of PE-treated cardiomyocytes corroborated this finding and indicated that CIP expression is regulated at the transcriptional level (Figure 6D). These results demonstrate that the expression of CIP is repressed in hypertrophic cardiomyocytes and hearts, in vitro and in vivo.CIP Represses Cardiomyocyte HypertrophyThese observations established an inverse correlation between the expression of CIP and cardiac hypertrophy and suggests that CIP could directly regulate cardiomyocyte hypertrophy. To test this hypothesis, we overexpressed CIP in cardiomyocytes by adenoviral gene transfer. Neonatal rat cardiomyocytes were isolated, cultured in vitro, and treated with PE to induce hypertrophy as previously described.51 PE treatment dramatically induced cardiomyocyte hypertrophy, as measured by increased cell surface area (Figure 6E). Overexpression of CIP did not result in significant change to cardiomyocytes. However, CIP significantly attenuated PE-induced hypertrophy (Figure 6E). Quantification of cardiomyocyte size confirmed the observation (Figure 6F). At the level of gene expression, PE induced expression of hypertrophic marker genes atrial natriuretic peptide, B-type natriuretic peptide, and β-myosin heavy chain, as expected. PE-induced expression of hypertrophic marker genes was inhibited by CIP (Figure 6G). Together, our results suggest that CIP negatively regulates cardiomyocyte hypertrophy.DiscussionIn the present study, we identified CIP as a novel cardiac-specific expressed nuclear protein. We demonstrated that CIP expression is restricted to cardiomyocytes. We further characterized CIP as a transcription cofactor of LIM/homeodomain transcription factor Isl1. We found that CIP physically interacts with Isl1 and represses its transcriptional activity. Functionally, we established that CIP represses cardiomyocyte hypertrophy.We identified Isl1 as a CIP-interacting protein via unbiased yeast two-hybrid screening and subsequently confirmed their physical and functional interactions. Our data suggest that a direct interaction between these two proteins is needed for CIP to repress the transactivity of Isl1. Isl1 is expressed in the second heart field and is required for the morphogenesis of embryonic heart.23 Interestingly, Isl1-positive progenitor cells were shown to be multi-potent and give rise to cardiac, smooth muscle, and endothelial cell lineages.22,52 Given that CIP interacts with Isl1 and is expressed in cardiac progenitors, we speculate that CIP might participate in the function of Isl1 in cell fate determination during early cardiac development. Despite its pivotal role in development, less is known about how Isl1 works at the molecular level. Isl1 is a LIM/homeodomain transcription factor and it synergistically activates insulin gene transcription with BETA2 in pancreatic beta cells.53 Furthermore, Isl1 forms a complex with Jak1 and Stat3 and triggers the tyrosine phosphorylation of Jak1 and its kinase activity.54 Isl1 works upstream of the sonic hedgehog signaling and the MEF2C transcription factor in the heart.21,46 Our data showed that the transcriptional activity of Isl1, although not very strong when compared with many other transcription factors, was repressed by CIP. This suggests another layer of regulation of this transcription factor.In addition to its unique cardiac expression pattern, several lines of evidences point to CIP as an important regulator of cardiac gene expression and cardiac hypertrophy. Intriguingly, our data showed the capability of CIP to repress Isl1-mediated activation of the MEF2C enhancer. It had been shown that MEF2C was upregulated during pathological hypertrophy.50 Transgenic mice expressing a dominant-negative MEF2C displayed attenuated postnatal growth of myocardium.48,49 Given the critical role of MEF2C in cardiac development and hypertrophy, it is reasonable to predict that CIP is also involved in this process. Consistent with this view, we found that the expression level of CIP was consistently downregulated in cardiac hypertrophy models, both in vivo and in vitro. Most importantly, we provide evidence to demonstrate that overexpression of CIP partially represses agonist-induced cardiomyocyte hypertrophy. It remains to be determined, using genetic approaches, whether CIP participates in the regulation of cardiac gene expression during heart development and whether dysregulation of CIP expression will lead to cardiac pathophysiological condition or human cardiovascular disease. It will also be important to understand how CIP participates in the regulatory pathways of cardiac hypertrophy.CIP is evolutionarily conserved in vertebrate. Its orthologs were found in things from fish to humans, suggesting it has conserved biological function and may play an important role in cardiac development and disease. CIP appears to stand as a single member of this class of protein, and no homologous protein is encoded in the mouse genome. Recently, another splice variant in the locus of 2310046A06Rik was identified as a lamin-interacting protein (MLIP).44 MLIP was shown to interact directly with lamin A/C (LMNA), a nuclear envelope protein. Interestingly, LMNA is known to interact with many transcription factors/cofactors, including retinoblastoma transcriptional regulator (RB),55 germ cell-less,56 sterol response element binding protein,57 C-Fos,58 and zinc finger protein 239.59 However, the functional significance of such interaction remains to be fully understood. LMNA is essential for the function of heart and human patients with LMNA mutation often exhibit adult-onset dilated cardiomyopathy accompanied with conduction-system disease.60 LMNA-null mice display severe skeletal muscle atrophy and dilated cardiomyopathy and die by 8 weeks.61,62 These observations suggest that the LMNA-interacting protein CIP/MLIP could be part of the network that regulates gene expression in LMNA-related dilated cardiomyopathy. It will be interesting to understand how CIP/MLIP modulates the function of LMNA and, most importantly, whether CIP/MLIP is directly related to human cardiac and muscle diseases.Our data demonstrate that CIP is expressed in the developing and adult heart. More specifically, we show that CIP expression is restricted to cardiomyocytes and cardiac progenitors during development. We have also observed transient CIP expression in the skeletal muscle. However, the level of CIP expression in skeletal muscle appears to be low when compared with that of cardiac muscle. Moreover, skeletal muscle expression of CIP is not detected at all stages of embryos or postnatal mice. It will be interesting to investigate whether CIP plays a role in skeletal muscle development and function.Database Searching and Cloning of CIPWe screened for novel cardiac-specific genes in silico by performing a Basic Local Alignment Search Tool search with expressed sequence tags from mouse embryonic heart cDNA libraries in the database as described previously.36 One of the cDNA sequences identified in this screen (access number AA919489) corresponded to the 3′ untranslated region of the CIP transcript and was found in a cDNA library of embryonic day 13.5 mouse embryonic hearts. This short cDNA fragment (273 nt) was used as a probe to screen a mouse embryonic heart cDNA library. Several cDNA clones were identified. Among them, the longest clone is 1336 nt in length. Polymerase chain reaction-based cloning was further used to identify additional cDNA isoforms.CIP Reporter MiceThe CIP reporter line with a retroviral gene trap cassette inserted into mouse genome chromosome 9 (between chromosome 9: 77 046 001 and 77 046 002) was generated and obtained from Texas A M Institute for Genomic Medicine. The gene trap cassette containing the selectable marker β-geo, a functional fusion between the β-galactosidase and neomycin resistance gene, was inserted into the putative third intron of the CIP gene (Online Figure III).In Situ Hybridization and Northern AnalysisWhole-mount and section in situ hybridization and Northern analyses were performed as described.36,63 The 273 nt cDNA fragment corresponding to the 3′ untranslated region of the CIP gene was used as a probe to perform both whole-mount and tissue section in in situ hybridization on staged mouse embryos.Constructs, Cell Culture, and Luciferase Reporter AssaysHela, COS7, and HEK293T cells were cultured in DMEM supplemented with 10% fetal bovine serum in a 5% CO2 atmosphere at 37°C. Luciferase reporter constructs fused with the MEF2C enhancer were as reported and are generous gifts of Dr. Brian Black (University of California, San Francisco).46 Transfections were performed with either FuGENE6 (Roche) or lipofectamine (Invitrogen) reagents according to manufacturer instructions. Unless otherwise indicated, 100 ng of reporter plasmid and 100 ng of each activator plasmid were applied; 48 hours after transfection, cell extracts were prepared and luciferase activity was determined. For luciferase assay, normalized luciferase expression from triplicate samples in 12-well plates relative to LacZ expression was calculated, and the results are expressed as fold activation over the value relative to the control (luciferase reporter and empty pcDNA). The CIP adenoviral expression construct contained full-length CIP cDNA and was constructed as previously described.51Tissue Dissociation and Cell Sorting of Mouse EmbryosThe embryonic day 10.5 embryonic hearts were isolated by microdissection and dissociated to single cells by collagenase digestion as previously described.64 Isolated cells were fluorescence-activated cell-sorted into green fluorescent protein-positive and green fluorescent protein-negative populations. Sorted cells were collected into Trizol (Invitrogen) and frozen at −20°C for RNA isolation.Immunochemistry and β-Galactosidase StainingStaged mouse embryos were dissected out, collected, and fixed in 4% paraformaldehyde at 4°C for 4 hours. After washing in phosphate-buffered saline, embryos were treated in 15% and 30% sucrose for 2 hours each and embedded in OCT. Approximately 5-μm to 8-μm cryostat sections were collected on positively charged slides. Sections were washed in phosphate-buffered saline, blocked in 5% serum/phosphate-buffered saline, and subjected to immunostaining. Antibody sources were as follows: green fluorescent protein (Invitrogen); platelet/endothelial cell adhesion molecule (BD Biosciences); Nkx2-5 (Santa Cruz); Wt1 (Santa Cruz); cardiac Troponin T (Tnnt2; generous gift of Dr. Jim Lin, University of Iowa); cardiac Troponin I (Tnni3; Santa Cruz); Actn2 (Sigma); Isl1 (DSHB; University of Iowa); and β-galatosidase (MP Biomedical). Alexa-488 and Alexa-594 secondary antibodies (Invitrogen). Fluorescently stained slides were counterstained with DAPI and imaged with an FV1000 confocal microscope (Olympus). For β-galactosidase staining, samples were stained with a solution containing 5 mmol/L potassium ferricyanide, 5 mmol/L potassium ferrocyanide, and 1 mg/mL X-gal substrate at 37°C for 12 hours after fixation.Quantitative Reverse-Transcriptase Polymerase Chain Reaction and Western Blot AnalysesTotal RNAs were isolated using Trizol reagent (Invitrogen) from cells and tissue samples. For quantitative reverse-transcriptase polymerase chain reaction, 2.0-μg RNA samples were reverse-transcribed to cDNA by using random hexamers and moloney murine leukemia virus reverse-transcriptase (Invitrogen) in a 20-μL reaction system. In each analysis, 0.1 μL cDNA pool was used for quantitative polymerase chain reaction. For Western blot analyses, cell extractions were cleared by 10 000g centrifugation for 10 minutes at 4°C. Samples were subsequently separated by SDS/PAGE and transferred to polyvinylidene fluoride membranes that were incubated with 5% milk and anti-FLAG (1:1000; Sigma), anti-Myc (1:1000; Santa Cruz), and β-tubulin (1:10 000; Sigma) overnight at 4°C and then washed three times with tris-buffered saline tween-20 buffer before adding secondary antibody. Polyclonal antibodies against the CIP protein were generated by immunizing rabbits with CIP proteins that were expressed and purified as GST-CIP fusion proteins in bacterial. Specific protein bands were visualized by using enhanced chemiluminescence (Invitrogen) reagents.Coimmunoprecipitation AssaysCOS7 cells were transiently transfected with plasmids encoding FLAG-tagged CIP and Myc-tagged Isl1 proteins with FuGENE6 (Roche). Cells were harvested 48 hours after transfection in lysis buffer composed of 1× phosphate-buffered saline containing 0.5% Triton X-100, 1 mmol/L EDTA, 1 mmol/L phenylmethylsulfonyl fluoride, 150 mmol/L of NaCl, and complete protease inhibitors (Roche). After a brief sonication and removal of cellular debris by centrifugation, FLAG-tagged CIP proteins were precipitated with anti-FLAG antibodies and protein A/G beads and associated proteins were analyzed by Western blotting with anti-Myc antibodies.In Vitro GST Protein-Binding AssaysPlasmids encoding a GST fusion with CIP were transformed into BL21 plus cells (Stratagene). The cells were grown at 37°C in 2× yeast-extract tryptone medium to an optical density of 1.0. Isopropyl-β-d-thiogalactopyranoside (50 μmol/L) was then added to the culture to induce protein expression. After being shaken at room temperature for 4 hours, the cells were harvested and the GST protein was purified with glutathione beads. Isl1 proteins translated in vitro were labeled with 35S methionine by using a TNT T7 reticulocyte lysate system (Promega). Glutathione beads conjugated with GST fusion protein were incubated with 10 μL of TNT product at 4°C for 2 hours in 500 μL of GST-binding buffer (20 mmol/L Tris, pH 7.3/150 mmol/L NaCl/0.5% Nonidet P-40/protease inhibitor/1 mmol/L phenylmethylsulfonyl fluoride). The beads were washed three times with GST-binding buffer; 50 μL of SDS loading buffer was then added to the beads. After boiling, 20 μL was loaded onto an SDS/PAGE gel and analyzed by autoradiography.Cardiomyocyte CultureNeonatal rat cardiomyocytes were prepared as previously described.51 Briefly, 18 hours after plating, cells were changed into serum-free medium and infected with adenovirus (Ad-GFP for control and Ad-CIP) at a multiplicity of infection of 25. Twenty-four hours later, cells were treated with hypertrophic agents PE. Cells were harvested 24 hours after PE treatment for RNA isolation or 48 hours after PE treatment for immunochemistry.Non-standard Abbreviations and AcronymsACTN2cardiac actinANFatrial natriuretic factorBNPbrain natriuretic peptidecDNAcomplementary DNACIPcardiac Isl1-interacting proteinETendothelinFACSfluorescence-activated cell sortingIPimmunoprecipitationIsl1Islet1LIFleukemia inhibitory factorLMNAlamin A/CLucluciferaseMEF2myocyte enhancer factor 2MYCmyosin heavy chainPEphenylephrineqPCRquantitative polymerase chain reactionTACtransverse aortic constrictionTNNT2cardiac troponin TUTRuntranslated regionWBWestern blotAcknowledgmentsThe authors thank members of the Wang laboratory for advice and support.Sources of FundingWork in Dr Wang\'s laboratory was supported by the March of Dimes Birth Defect Foundation and National Institutes of Health. Z. P. Huang is a postdoctoral fellow and D. Z. Wang is an Established Investigator of the American Heart Association.DisclosuresNone.FootnotesIn December 2011, the average time from submission to first decision for all original research papers submitted to Circulation Research was 14.29 days.The online-only Data Supplement is available with this article at http://circres.ahajournals.org/lookup/suppl/doi:10.1161/CIRCRESAHA.111.259663/-/DC1.Correspondence to Da-Zhi Wang, PhD, Children\'s Hospital Boston, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115. E-mail [email protected]tch.harvard.eduReferences1. Olson EN.A decade of discoveries in cardiac biology.Nat Med.2004;467–474.CrossrefMedlineGoogle Scholar2. Musunuru K, Domian IJ, Chien KR.Stem cell models of cardiac development and disease.Annu Rev Cell Dev Biol.667–687.CrossrefMedlineGoogle Scholar3. Komuro I, Izumo S.Csx: a murine homeobox-containing gene specifically expressed in the developing heart.Proc Natl Acad Sci U S A.1993;8145–8149.CrossrefMedlineGoogle Scholar4. Lyons I, Parsons LM, Hartley L, Li R, Andrews JE, Robb L, Harvey RP.Myogenic and morphogenetic defects in the heart tubes of murine embryos lacking the homeo box gene Nkx2-5.Genes Dev.1995;1654–1666.CrossrefMedlineGoogle Scholar5. Lints TJ, Parsons LM, Hartley L, Lyons I, Harvey RP.Nkx-2.5: a novel murine homeobox gene expressed in early heart progenitor cells and their myogenic descendants.Development.1993; 119:969.MedlineGoogle Scholar6. Lin Q, Schwarz J, Bucana C, Olson EN.Control of mouse cardiac morphogenesis and myogenesis by transcription factor MEF2C.Science.1997; 276:1404–1407.CrossrefMedlineGoogle Scholar7. Lilly B, Zhao B, Ranganayakulu G, Paterson BM, Schulz RA, Olson EN.Requirement of MADS domain transcription factor D-MEF2 for muscle formation in Drosophila.Science.1995; 267:688–693.CrossrefMedlineGoogle Scholar8. Edmondson DG, Lyons GE, Martin JF, Olson EN.Mef2 gene expression marks the cardiac and skeletal muscle lineages during mouse embryogenesis.Development.1994; 120:1251–1263.MedlineGoogle Scholar9. Creemers EE, Sutherland LB, Oh J, Barbosa AC, Olson EN.Coactivation of MEF2 by the SAP domain proteins myocardin and MASTR.Mol Cell.2006;83–96.CrossrefMedlineGoogle Scholar10. Naya FJ, Olson E.MEF2: a transcriptional target for signaling pathways controlling skeletal muscle growth and differentiation.Curr Opin Cell Biol.1999;683–688.CrossrefMedlineGoogle Scholar11. Black BL, Olson EN.Transcriptional control of muscle development by myocyte enhancer factor-2 (MEF2) proteins.Annu Rev Cell Dev Biol.1998;167–196.CrossrefMedlineGoogle Scholar12. Molkentin JD, Kalvakolanu DV, Markham BE.Transcription factor GATA-4 regulates cardiac muscle-specific expression of the alpha-myosin heavy-chain gene.Mol Cell Biol.1994;4947–4957.CrossrefMedlineGoogle Scholar13. Peterkin T, Gibson A, Loose M, Patient R.The roles of GATA-4, -5 and -6 in vertebrate heart development.Semin Cell Dev Biol.2005;83–94.CrossrefMedlineGoogle Scholar14. Burch JB.Regulation of GATA gene expression during vertebrate development.Semin Cell Dev Biol.2005;71–81.CrossrefMedlineGoogle Scholar15. Abu-Issa R, Kirby ML.Heart field: from mesoderm to heart tube.Annu Rev Cell Dev Biol.2007;45–68.CrossrefMedlineGoogle Scholar16. Harvey RP, Meilhac SM, Buckingham ME.Landmarks and lineages in the developing heart.Circ Res.2009; 104:1235–1237.LinkGoogle Scholar17. Kelly RG, Buckingham ME.The anterior heart-forming field: voyage to the arterial pole of the heart.Trends Genet.2002;210–216.CrossrefMedlineGoogle Scholar18. Black BL.Transcriptional pathways in second heart field development.Semin Cell Dev Biol.2007;67–76.CrossrefMedlineGoogle Scholar19. Rochais F, Mesbah K, Kelly RG.Signaling pathways controlling second heart field development.Circ Res.2009; 104:933–942.LinkGoogle Scholar20. Vincent SD, Buckingham ME.How to make a heart: the origin and regulation of cardiac progenitor cells.Curr Top Dev Biol.2010;1–41.CrossrefMedlineGoogle Scholar21. Lin L, Bu L, Cai CL, Zhang X, Evans S.Isl1 is upstream of sonic hedgehog in a pathway required for cardiac morphogenesis.Dev Biol.2006; 295:756–763.CrossrefMedlineGoogle Scholar22. Laugwitz KL, Moretti A, Lam J, Gruber P, Chen Y, Woodard S, Lin LZ, Cai CL, Lu MM, Reth M, Platoshyn O, Yuan JX, Evans S, Chien KR.Postnatal isl1+ cardioblasts enter fully differentiated cardiomyocyte lineages.Nature.2005; 433:647–653.CrossrefMedlineGoogle Scholar23. Cai CL, Liang X, Shi Y, Chu PH, Pfaff SL, Chen J, Evans S.Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart.Dev Cell.2003;877–889.CrossrefMedlineGoogle Scholar24. Mann T, Bodmer R, Pandur P.The Drosophila homolog of vertebrate Islet1 is a key component in early cardiogenesis.Development.2009; 136:317–326.CrossrefMedlineGoogle Scholar25. Ahmad F, Arad M, Musi N, He H, Wolf C, Branco D, Perez-Atayde AR, Stapleton D, Bali D, Xing Y, Tian R, Goodyear LJ, Berul CI, Ingwall JS, Seidman CE, Seidman JG.Increased alpha2 subunit-associated AMPK activity and PRKAG2 cardiomyopathy.Circulation.2005; 112:3140–3148.LinkGoogle Scholar26. Frey N, Olson EN.Cardiac hypertrophy: the good, the bad, and the ugly.Annu Rev Physiol.2003;45–79.CrossrefMedlineGoogle Scholar27. Molkentin JD, Lu JR, Antos CL, Markham B, Richardson J, Robbins J, Grant SR, Olson EN.A calcineurin-dependent transcriptional pathway for cardiac hypertrophy.Cell.1998;215–228.CrossrefMedlineGoogle Scholar28. Seidman JG, Seidman C.The genetic basis for cardiomyopathy: from mutation identification to mechanistic paradigms.Cell.2001; 104:557–567.CrossrefMedlineGoogle Scholar29. Heineke J, Molkentin JD.Regulation of cardiac hypertrophy by intracellular signalling pathways.Nat Rev Mol Cell Biol.2006;589–600.CrossrefMedlineGoogle Scholar30. Rose BA, Force T, Wang Y.Mitogen-activated protein kinase signaling in the heart: angels versus demons in a heart-breaking tale.Physiol Rev.2010;1507–1546.CrossrefMedlineGoogle Scholar31. Jaalouk DE, Lammerding J.Mechanotransduction gone awry.Nat Rev Mol Cell Biol.2009;63–73.CrossrefMedlineGoogle Scholar32. Zhang CL, McKinsey TA, Chang S, Antos CL, Hill JA, Olson EN.Class II histone deacetylases act as signal-responsive repressors of cardiac hypertrophy.Cell.2002; 110:479–488.CrossrefMedlineGoogle Scholar33. Haberland M, Montgomery RL, Olson EN.The many roles of histone deacetylases in development and physiology: implications for disease and therapy.Nat Rev Genet.2009;32–42.CrossrefMedlineGoogle Scholar34. McKinsey TA, Olson EN.Toward transcriptional therapies for the failing heart: chemical screens to modulate genes.J Clin Invest.2005; 115:538–546.CrossrefMedlineGoogle Scholar35. Hauschka SD.Myocardin. a novel potentiator of SRF-mediated transcription in cardiac muscle.Mol Cell.2001;1–2.CrossrefMedlineGoogle Scholar36. Wang D, Chang PS, Wang Z, Sutherland L, Richardson JA, Small E, Krieg PA, Olson EN.Activation of cardiac gene expression by myocardin, a transcriptional cofactor for serum response factor.Cell.2001; 105:851–862.CrossrefMedlineGoogle Scholar37. Pipes GC, Creemers EE, Olson EN.The myocardin family of transcriptional coactivators: versatile regulators of cell growth, migration, and myogenesis.Genes Dev.2006;1545–1556.CrossrefMedlineGoogle Scholar38. Wang DZ, Olson EN.Control of smooth muscle development by the myocardin family of transcriptional coactivators.Curr Opin Genet Dev.2004;558–566.CrossrefMedlineGoogle Scholar39. Wang Z, Wang DZ, Hockemeyer D, McAnally J, Nordheim A, Olson EN.Myocardin and ternary complex factors compete for SRF to control smooth muscle gene expression.Nature.2004; 428:185–189.CrossrefMedlineGoogle Scholar40. Wang Z, Wang DZ, Pipes GC, Olson EN.Myocardin is a master regulator of smooth muscle gene expression.Proc Natl Acad Sci U S A.2003; 100:7129–7134.CrossrefMedlineGoogle Scholar41. Li S, Wang DZ, Wang Z, Richardson JA, Olson EN.The serum response factor coactivator myocardin is required for vascular smooth muscle development.Proc Natl Acad Sci U S A.2003; 100:9366–9370.CrossrefMedlineGoogle Scholar42. Li J, Zhu X, Chen M, Cheng L, Zhou D, Lu MM, Du K, Epstein JA, Parmacek MS.Myocardin-related transcription factor B is required in cardiac neural crest for smooth muscle differentiation and cardiovascular development.Proc Natl Acad Sci U S A.2005; 102:8916–8921.CrossrefMedlineGoogle Scholar43. Oh J, Richardson JA, Olson EN.Requirement of myocardin-related transcription factor-B for remodeling of branchial arch arteries and smooth muscle differentiation.Proc Natl Acad Sci U S A.2005; 102:15122–15127.CrossrefMedlineGoogle Scholar44. Ahmady E, Deeke SA, Rabaa S, Kouri L, Kenney L, Stewart AF, Burgon PG.Identification of a novel muscle A-type lamin-interacting protein (MLIP).J Biol Chem. 286:19702–19713.CrossrefMedlineGoogle Scholar45. Pfaff SL, Mendelsohn M, Stewart CL, Edlund T, Jessell TM.Requirement for LIM homeobox gene Isl1 in motor neuron generation reveals a motor neuron-dependent step in interneuron differentiation.Cell.1996;309–320.CrossrefMedlineGoogle Scholar46. Dodou E, Verzi MP, Anderson JP, Xu SM, Black BL.Mef2c is a direct transcriptional target of ISL1 and GATA factors in the anterior heart field during mouse embryonic development.Development.2004; 131:3931–3942.CrossrefMedlineGoogle Scholar47. Passier R, Zeng H, Frey N, Naya FJ, Nicol RL, McKinsey TA, Overbeek P, Richardson JA, Grant SR, Olson EN.CaM kinase signaling induces cardiac hypertrophy and activates the MEF2 transcription factor in vivo.J Clin Invest.2000; 105:1395–1406.CrossrefMedlineGoogle Scholar48. Xu J, Gong NL, Bodi I, Aronow BJ, Backx PH, Molkentin JD.Myocyte enhancer factors 2A and 2C induce dilated cardiomyopathy in transgenic mice.J Biol Chem.2006; 281:9152–9162.CrossrefMedlineGoogle Scholar49. van Oort RJ, van Rooij E, Bourajjaj M, Schimmel J, Jansen MA, van der Nagel R, Doevendans PA, Schneider MD, van Echteld CJ, De Windt LJ.MEF2 activates a genetic program promoting chamber dilation and contractile dysfunction in calcineurin-induced heart failure.Circulation.2006; 114:298–308.LinkGoogle Scholar50. Kolodziejczyk SM, Wang L, Balazsi K, DeRepentigny Y, Kothary R, Megeney LA.MEF2 is upregulated during cardiac hypertrophy and is required for normal post-natal growth of the myocardium.Curr Biol.1999;1203–1206.CrossrefMedlineGoogle Scholar51. Xing W, Zhang TC, Cao D, Wang Z, Antos CL, Li S, Wang Y, Olson EN, Wang DZ.Myocardin induces cardiomyocyte hypertrophy.Circ Res.2006;1089–1097.LinkGoogle Scholar52. Moretti A, Caron L, Nakano A, Lam JT, Bernshausen A, Chen Y, Qyang Y, Bu L, Sasaki M, Martin-Puig S, Sun Y, Evans SM, Laugwitz KL, Chien KR.Multipotent embryonic isl1+ progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification.Cell.2006; 127:1151–1165.CrossrefMedlineGoogle Scholar53. Zhang H, Wang WP, Guo T, Yang JC, Chen P, Ma KT, Guan YF, Zhou CY.The LIM-homeodomain protein ISL1 activates insulin gene promoter directly through synergy with BETA2.J Mol Biol.2009; 392:566–577.CrossrefMedlineGoogle Scholar54. Hao A, Novotny-Diermayr V, Bian W, Lin B, Lim CP, Jing N, Cao X.The LIM/homeodomain protein Islet1 recruits Janus tyrosine kinases and signal transducer and activator of transcription 3 and stimulates their activities.Mol Biol Cell.2005;1569–1583.CrossrefMedlineGoogle Scholar55. Johnson BR, Nitta RT, Frock RL, Mounkes L, Barbie DA, Stewart CL, Harlow E, Kennedy BK.A-type lamins regulate retinoblastoma protein function by promoting subnuclear localization and preventing proteasomal degradation.Proc Natl Acad Sci U S A.2004; 101:9677–9682.CrossrefMedlineGoogle Scholar56. Kimura T, Ito C, Watanabe S, Takahashi T, Ikawa M, Yomogida K, Fujita Y, Ikeuchi M, Asada N, Matsumiya K, Okuyama A, Okabe M, Toshimori K, Nakano T.Mouse germ cell-less as an essential component for nuclear integrity.Mol Cell Biol.2003;1304–1315.CrossrefMedlineGoogle Scholar57. Lloyd DJ, Trembath RC, Shackleton S.A novel interaction between lamin A and SREBP1: implications for partial lipodystrophy and other laminopathies.Hum Mol Genet.2002;769–777.CrossrefMedlineGoogle Scholar58. Ivorra C, Kubicek M, Gonzalez JM, Sanz-Gonzalez SM, Alvarez-Barrientos A, O\'Connor JE, Burke B, Andres V.A mechanism of AP-1 suppression through interaction of c-Fos with lamin A/C.Genes Dev.2006;307–320.CrossrefMedlineGoogle Scholar59. Dreuillet C, Tillit J, Kress M, Ernoult-Lange M.In vivo and in vitro interaction between human transcription factor MOK2 and nuclear lamin A/C.Nucleic Acids Res.2002;4634–4642.CrossrefMedlineGoogle Scholar60. Sylvius N, Tesson F.Lamin A/C and cardiac diseases.Curr Opin Cardiol.2006;159–165.CrossrefMedlineGoogle Scholar61. Ostlund C, Bonne G, Schwartz K, Worman HJ.Properties of lamin A mutants found in Emery-Dreifuss muscular dystrophy, cardiomyopathy and Dunnigan-type partial lipodystrophy.J Cell Sci.2001; 114(Pt 24):4435–4445.MedlineGoogle Scholar62. Raharjo WH, Enarson P, Sullivan T, Stewart CL, Burke B.Nuclear envelope defects associated with LMNA mutations cause dilated cardiomyopathy and Emery-Dreifuss muscular dystrophy.J Cell Sci.2001; 114(Pt 24):4447–4457.MedlineGoogle Scholar63. Wang DZ, Reiter RS, Lin JL, Wang Q, Williams HS, Krob SL, Schultheiss TM, Evans S, Lin JJ.Requirement of a novel gene, Xin, in cardiac morphogenesis.Development.1999; 126:1281–1294.MedlineGoogle Scholar64. Zhou B, Ma Q, Rajagopal S, Wu SM, Domian I, Rivera-Feliciano J, Jiang D, von Gise A, Ikeda S, Chien KR, Pu WT.Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart.Nature.2008; 454:109–113.CrossrefMedlineGoogle ScholarNovelty and SignificanceWhat Is Known?Cardiac hypertrophy is one of the most common responses of the heart in response to mechanical stress and pathological stimuli.Chronic activation of pathological hypertrophy often leads to heart failure and sudden death.Several signaling pathways and transcriptional networks activate cardiac hypertrophy.Mechanisms underlying the repression of hypertrophy are not well-understood.What New Information Does This Article Contribute?This study identified cardiac Isl1-interacting protein (CIP) as a novel cardiac-specific nuclear protein.CIP is dynamically regulated in response to cardiac hypertrophy.Overexpression of CIP represses cardiomyocyte hypertrophy.Congestive heart failure is a common outcome of a variety of primary cardiovascular disease entities, including coronary artery disease, and hypertension,. Nevertheless, mechanisms underlying the progression of compensated hypertrophy to heart failure remain unclear. Although the pathophysiology of cardiac hypertrophy has been extensively studied and much is known about signaling pathways that activate cardiac hypertrophy, processes leading to the repression of cardiac hypertrophy are less well understood. In this study, we identify CIP as a novel gene expressed in embryonic and adult hearts. Combinational approaches demonstrated that CIP expression is limited to cardiomyocytes, and that it is no expressed in other cell types (fibroblast, endothelial cells or vascular smooth muscle cells) of the heart. Our results show that CIP physically interacts with Isl1, a transcription factor essential for heart development and the fate of cardiac progenitors. Functionally, we found that CIP is a transcriptional repressor that represses agonist-induced cardiomyocyte hypertrophy and the expression of hypertrophic marker genes. These findings suggest that CIP may be a potential target in the development of future therapies for cardiac hypertrophy and heart failure. Manuscript receivedOctober 25, 2011Manuscript acceptedJanuary 27, 2011Originally publishedFebruary 16, 2012

本文链接: http://rdsystems.immuno-online.com/view-66816.html

发布于 : 2024-05-09 阅读()
品牌分类
链霉亲和素研究产品 前列腺组织微阵列 IL8研究产品 INSTA-Blot结肠组织肿瘤学WB膜 HLA-A研究产品 VEGF 165研究产品 VECTASTAIN ABC包 乳腺片 细胞内染色试剂盒
GRP75/HSPA9B/凡人药盒 IL-17/IL-17A试剂盒 ChABC抗体对 IgE抗体 小肠空肠玻片 RunBlue BIS-TRIS凝胶12%-10x10cm Novex兼容支持产品 HIF-1α抗体包 肺组织微阵列 肝脏研究产品 脾片 RunBlue BIS-TRIS凝胶4-12%-10x10cm Novex兼容支持产品 高强度脯氨酸羟化酶抗体包 TLR4抑制剂 pCL生态逆转录病毒包装载体支持产品 HBeAg-ELISA试剂盒 聚肌苷酸支持产品 OSMR裂解物 Lightning-Link合包 QuikChIP(带装载控制)套件 VECTASTAIN精英ABC包 RunBlue TEO Tricine SDS凝胶16%-10x10cm Novex兼容支持产品 输尿管玻片 INSTA-Blot检测男性肺组织癌细胞对WB膜 Pam3CSK4、TLR1和TLR2配体支持产品 乳腺组织芯片 pSIVA凋亡检测试剂盒 NOXA1抗体对 胃研究产品 纹状体研究产品 多组织微阵列 RunBlue TEO Tricine SDS蛋白质凝胶12%-10x10 Novex兼容研究产品 FlexLISA Phosphatase Alkaline睡袋 RunBlue TEO Tricine SDS凝胶10%-8x10cm Bio-Rad兼容支持产品 人类调节性T细胞研究产品 PiColorLock磷酸检测试剂产品的支持 多器官研究产品 人造血祖细胞试剂盒 v6.5小鼠胚胎干细胞支持产品 BMP7研究产品 格列本脲抑制剂 人骨髓间充质干细胞研究产品 DAPI解决方案支持产品 Lightning-Link R-PE包 α-平滑肌肌动蛋白抗体对 MMP8研究产品 聚乙二醇抗体对 NFkB p50(NLS)抑制剂 SR-BI抗体包 ATM研究产品 小鼠红细胞溶解试剂盒 FGF碱性/FGF2/bFGF蛋白 IRE1α抗体包 多组织微阵列 RunBlue BIS-TRIS凝胶8%-10x10cm Novex兼容支持产品 扁桃体片 CpG寡核苷酸支持产品 肝组织微阵列 CACNA1A蛋白 MALP-2研究产品 淋巴结切片 CEACAM18研究产品 肾组织微阵列 PD1研究产品 pCL文昌鱼逆转录病毒包装载体支持产品 RunBlue TEO Tricine SDS凝胶20%-10x10 Novex兼容支持产品 CD20研究产品 S100A9研究产品 胰腺切片 IgG1抗体 MOSPD3裂解物 润蓝预拉伸分子量标记三色支撑产品 CCL20/MIP-3α试剂盒 INSTA-Blot人细胞系WB膜 IgA抗体 小脑载玻片 INSTA-Blot乳腺癌细胞膜 HIF-2α/EPAS1抗体包 papplaysin-1/PAPP-A蛋白 RunBlue TEO Tricine SDS凝胶4-20%-8x10cm生物放射性兼容支持产品 小鼠间充质干细胞试剂盒 mGluR3裂解物 OIP5抗体对 人红细胞溶解试剂盒 咪喹莫特,TLR7配体支持产物 CpG寡核苷酸小鼠支持产品 IKKγ抑制剂 GDC蛋白 多能干细胞转录因子抗体包 CA15-3elisa试剂盒 盐酸右芬氟拉明抑制剂 TSLP套件 胶原VIα1蛋白 Ki67/MKI67抗体 CD8β蛋白 免疫球蛋白(H L)抗体 INSTA-Blot法检测女性肺组织癌细胞对WB膜 单胺氧化酶研究产品 润蓝TEO Tricine SDS凝胶4-8%-8x10cm Bio-Rad兼容支持产品 pCL-10A1逆转录病毒包装载体支持产品 小鼠TH1细胞试剂盒 IgG-Fc抗体 胎盘片 RunBlue TEO Tricine SDS凝胶12%-8x10cm Bio-Rad兼容支持产品 色差芯片套件 结肠切片 INSTA-Blot大鼠组织WB膜 RunBlue TEO Tricine SDS凝胶16%-8x10cm Bio-Rad兼容支持产品 DGCR8基因敲除小鼠胚胎干细胞支持物 Lightning-Link APC包 RunBlue BIS-TRIS凝胶10%-10x10cm Novex兼容支持产品 ATPase试剂盒 自噬抗体包 INSTA-Blot人脑白细胞膜 RunBlue TEO Tricine SDS凝胶10%-10x10cm Novex兼容支持产品 INSTA杂交卵巢组织癌细胞对WB膜 PINK1研究产品 MyD88抑制剂 卵巢组织微阵列 Pam3CSK4、TLR1和TLR2配体支持产品 INSTA-Blot小鼠组织WB膜 氟橡胶支架产品 INSTA-Blot人组织WB膜 MMP-12试剂盒 血管生成素-1 ELISA试剂盒 RunBlue TEO Tricine SDS凝胶20%-10x10cm Novex兼容支持产品 gp96/HSP90B1/GRP94套件 HO-1/HMOX1/HSP32试剂盒 RelA/NFkB p65抑制剂 TIRAP(TLR2和TLR4)抑制剂 APE1氧化还原抑制剂 肾片 CD8蛋白 H/M多能干细胞试剂盒 血浆激肽释放酶肽和蛋白质 酵母多糖,TLR2配体支持产物 INSTA-Blot多物种脑、睾丸和卵巢WB膜 固定和渗透试剂盒 内质网应激/UPR研究产品 肺片 RunBlue TEO Tricine SDS凝胶8%-8x10cm Bio-Rad兼容支持产品 MUC1抗体对 FlexLISA合包 胶原蛋白II研究产品
显示分类
联络我们
服务热线:4000-520-616
(限工作日9:00-18:00)
QQ :1570468124
手机:18915418616