1
|
Srivastava Y, Donta M, Mireles LL, Paulucci-Holthauzen A, Shi L, Bedford MT, Waxham MN, McCrea PD. Exploring the PDZ, DUF, and LIM Domains of Pdlim5 in Dendrite Branching. Int J Mol Sci 2024; 25:8326. [PMID: 39125895 PMCID: PMC11312917 DOI: 10.3390/ijms25158326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/10/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
The branched architecture of neuronal dendrites is a key factor in how neurons form ordered networks and discoveries continue to be made identifying proteins and protein-protein interactions that direct or execute the branching and extension of dendrites. Our prior work showed that the molecular scaffold Pdlim5 and delta-catenin, in conjunction, are two proteins that help regulate the branching and elongation of dendrites in cultured hippocampal neurons and do so through a phosphorylation-dependent mechanism triggered by upstream glutamate signaling. In this report we have focused on Pdlim5's multiple scaffolding domains and how each contributes to dendrite branching. The three identified regions within Pdlim5 are the PDZ, DUF, and a trio of LIM domains; however, unresolved is the intra-molecular conformation of Pdlim5 as well as which domains are essential to regulate dendritic branching. We address Pdlim5's structure and function by examining the role of each of the domains individually and using deletion mutants in the context of the full-length protein. Results using primary hippocampal neurons reveal that the Pdlim5 DUF domain plays a dominant role in increasing dendritic branching. Neither the PDZ domain nor the LIM domains alone support increased branching. The central role of the DUF domain was confirmed using deletion mutants in the context of full-length Pdlim5. Guided by molecular modeling, additional domain mapping studies showed that the C-terminal LIM domain forms a stable interaction with the N-terminal PDZ domain, and we identified key amino acid residues at the interface of each domain that are needed for this interaction. We posit that the central DUF domain of Pdlim5 may be subject to modulation in the context of the full-length protein by the intra-molecular interaction between the N-terminal PDZ and C-terminal LIM domains. Overall, our studies reveal a novel mechanism for the regulation of Pdlim5's function in the regulation of neuronal branching and highlight the critical role of the DUF domain in mediating these effects.
Collapse
Affiliation(s)
- Yogesh Srivastava
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maxsam Donta
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Program in Genetics & Epigenetics, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
| | - Lydia L. Mireles
- Department of Neurobiology & Anatomy, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
| | | | - Leilei Shi
- Department of Epigenetics & Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mark T. Bedford
- Program in Genetics & Epigenetics, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
- Department of Epigenetics & Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - M. Neal Waxham
- Department of Neurobiology & Anatomy, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
- Program in Neuroscience, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
| | - Pierre D. McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Program in Genetics & Epigenetics, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
- Program in Neuroscience, University of Texas MD Anderson Cancer Center, UT Health GSBS, Houston, TX 77030, USA
| |
Collapse
|
2
|
Fu Y, Li S, Nie J, Yan D, Zhang B, Hao X, Zhang H. Expression of PDLIM5 Spliceosomes and Regulatory Functions on Myogenesis in Pigs. Cells 2024; 13:720. [PMID: 38667334 PMCID: PMC11049100 DOI: 10.3390/cells13080720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Meat yield, determined by muscle growth and development, is an important economic trait for the swine industry and a focus of research in animal genetics and breeding. PDZ and LIM domain 5 (PDLIM5) are cytoskeleton-related proteins that play key roles in various tissues and cells. These proteins have multiple isoforms, primarily categorized as short (PDLIM5-short) and long (PDLIM5-long) types, distinguished by the absence and presence of an LIM domain, respectively. However, the expression patterns of swine PDLIM5 isoforms and their regulation during porcine skeletal muscle development remain largely unexplored. We observed that PDLIM5-long was expressed at very low levels in pig muscles and that PDLIM5-short and total PDLIM5 were highly expressed in the muscles of slow-growing pigs, suggesting that PDLIM5-short, the dominant transcript in pigs, is associated with a slow rate of muscle growth. PDLIM5-short suppressed myoblast proliferation and myogenic differentiation in vitro. We also identified two single nucleotide polymorphisms (-258 A > T and -191 T > G) in the 5' flanking region of PDLIM5, which influenced the activity of the promoter and were associated with muscle growth rate in pigs. In summary, we demonstrated that PDLIM5-short negatively regulates myoblast proliferation and differentiation, providing a theoretical basis for improving pig breeding programs.
Collapse
Affiliation(s)
- Yu Fu
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| | - Shixin Li
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| | - Jingru Nie
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| | - Dawei Yan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China;
| | - Bo Zhang
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| | - Xin Hao
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| | - Hao Zhang
- National Engineering Laboratory for Livestock and Poultry Breeding, Beijing Key Laboratory of Animal Genetic Engineering, China Agricultural University, Beijing 100193, China; (Y.F.); (S.L.); (J.N.); (B.Z.)
| |
Collapse
|
3
|
Srivastava Y, Donta M, Mireles LL, Paulucci-Holthauzen A, Waxham MN, McCrea PD. Role of a Pdlim5:PalmD complex in directing dendrite morphology. Front Cell Neurosci 2024; 18:1315941. [PMID: 38414752 PMCID: PMC10896979 DOI: 10.3389/fncel.2024.1315941] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/18/2024] [Indexed: 02/29/2024] Open
Abstract
Neuronal connectivity is regulated during normal brain development with the arrangement of spines and synapses being dependent on the morphology of dendrites. Further, in multiple neurodevelopmental and aging disorders, disruptions of dendrite formation or shaping is associated with atypical neuronal connectivity. We showed previously that Pdlim5 binds delta-catenin and promotes dendrite branching. We report here that Pdlim5 interacts with PalmD, a protein previously suggested by others to interact with the cytoskeleton (e.g., via adducin/spectrin) and to regulate membrane shaping. Functionally, the knockdown of PalmD or Pdlim5 in rat primary hippocampal neurons dramatically reduces branching and conversely, PalmD exogenous expression promotes dendrite branching as does Pdlim5. Further, we show that each proteins' effects are dependent on the presence of the other. In summary, using primary rat hippocampal neurons we reveal the contributions of a novel Pdlim5:PalmD protein complex, composed of functionally inter-dependent components responsible for shaping neuronal dendrites.
Collapse
Affiliation(s)
- Yogesh Srivastava
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maxsam Donta
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Program in Genetics and Epigenetics, University of Texas MD Anderson Cancer Center UT Health GSBS, Houston, TX, United States
| | - Lydia L. Mireles
- Department of Neurobiology and Anatomy, UTHealth, Houston, TX, United States
| | | | - M. Neal Waxham
- Department of Neurobiology and Anatomy, UTHealth, Houston, TX, United States
- Program in Neuroscience, University of Texas MD Anderson Cancer Center UT Health GSBS, Houston, TX, United States
| | - Pierre D. McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Program in Genetics and Epigenetics, University of Texas MD Anderson Cancer Center UT Health GSBS, Houston, TX, United States
- Program in Neuroscience, University of Texas MD Anderson Cancer Center UT Health GSBS, Houston, TX, United States
| |
Collapse
|
4
|
Srivastava Y, Donta M, Mireles LL, Paulucci-Holthauzen A, Waxham MN, McCrea PD. Role of a Pdlim5:PalmD complex in directing dendrite morphology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.22.553334. [PMID: 37662414 PMCID: PMC10473622 DOI: 10.1101/2023.08.22.553334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Neuronal connectivity is regulated during normal brain development with the arrangement of spines and synapses being dependent on the morphology of dendrites. Further, in multiple neurodevelopmental and aging disorders, disruptions of dendrite formation or shaping is associated with atypical neuronal connectivity. We showed previously that Pdlim5 binds delta-catenin and promotes dendrite branching (Baumert et al., J Cell Biol 2020). We report here that Pdlim5 interacts with PalmD, a protein previously suggested by others to interact with the cytoskeleton (e.g., via adducin/ spectrin) and to regulate membrane shaping. Functionally, the knockdown of PalmD or Pdlim5 in rat primary hippocampal neurons dramatically reduces branching and conversely, PalmD exogenous expression promotes dendrite branching as does Pdlim5. Further, we show that effects of each protein are dependent on the presence of the other. In summary, using primary rat hippocampal neurons we reveal the contributions of a novel Pdlim5:PalmD protein complex, composed of functionally inter-dependent components responsible for shaping neuronal dendrites.
Collapse
|
5
|
Fisher LAB, Schöck F. The unexpected versatility of ALP/Enigma family proteins. Front Cell Dev Biol 2022; 10:963608. [PMID: 36531944 PMCID: PMC9751615 DOI: 10.3389/fcell.2022.963608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022] Open
Abstract
One of the most intriguing features of multicellular animals is their ability to move. On a cellular level, this is accomplished by the rearrangement and reorganization of the cytoskeleton, a dynamic network of filamentous proteins which provides stability and structure in a stationary context, but also facilitates directed movement by contracting. The ALP/Enigma family proteins are a diverse group of docking proteins found in numerous cellular milieus and facilitate these processes among others. In vertebrates, they are characterized by having a PDZ domain in combination with one or three LIM domains. The family is comprised of CLP-36 (PDLIM1), Mystique (PDLIM2), ALP (PDLIM3), RIL (PDLIM4), ENH (PDLIM5), ZASP (PDLIM6), and Enigma (PDLIM7). In this review, we will outline the evolution and function of their protein domains which confers their versatility. Additionally, we highlight their role in different cellular environments, focusing specifically on recent advances in muscle research using Drosophila as a model organism. Finally, we show the relevance of this protein family to human myopathies and the development of muscle-related diseases.
Collapse
|
6
|
Insulin and Insulin-Like Growth Factor 1 Signaling Preserves Sarcomere Integrity in the Adult Heart. Mol Cell Biol 2022; 42:e0016322. [PMID: 36125265 PMCID: PMC9583714 DOI: 10.1128/mcb.00163-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Insulin and insulin-like growth factor 1 (IGF1) signaling is transduced by insulin receptor substrate 1 (IRS1) and IRS2. To elucidate physiological and redundant roles of insulin and IGF1 signaling in adult hearts, we generated mice with inducible cardiomyocyte-specific deletion of insulin and IGF1 receptors or IRS1 and IRS2. Both models developed dilated cardiomyopathy, and most mice died by 8 weeks post-gene deletion. Heart failure was characterized by cardiomyocyte loss and disarray, increased proapoptotic signaling, and increased autophagy. Suppression of autophagy by activating mTOR signaling did not prevent heart failure. Transcriptional profiling revealed reduced serum response factor (SRF) transcriptional activity and decreased mRNA levels of genes encoding sarcomere and gap junction proteins as early as 3 days post-gene deletion, in concert with ultrastructural evidence of sarcomere disruption and intercalated discs within 1 week after gene deletion. These data confirm conserved roles for constitutive insulin and IGF1 signaling in suppressing autophagic and apoptotic signaling in the adult heart. The present study also identifies an unexpected role for insulin and IGF1 signaling in regulating an SRF-mediated transcriptional program, which maintains expression of genes encoding proteins that support sarcomere integrity in the adult heart, reduction of which results in rapid development of heart failure.
Collapse
|
7
|
Gan P, Wang Z, Morales MG, Zhang Y, Bassel-Duby R, Liu N, Olson EN. RBPMS is an RNA-binding protein that mediates cardiomyocyte binucleation and cardiovascular development. Dev Cell 2022; 57:959-973.e7. [PMID: 35472321 PMCID: PMC9116735 DOI: 10.1016/j.devcel.2022.03.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/04/2022] [Accepted: 03/25/2022] [Indexed: 12/13/2022]
Abstract
Noncompaction cardiomyopathy is a common congenital cardiac disorder associated with abnormal ventricular cardiomyocyte trabeculation and impaired pump function. The genetic basis and underlying mechanisms of this disorder remain elusive. We show that the genetic deletion of RNA-binding protein with multiple splicing (Rbpms), an uncharacterized RNA-binding factor, causes perinatal lethality in mice due to congenital cardiovascular defects. The loss of Rbpms causes premature onset of cardiomyocyte binucleation and cell cycle arrest during development. Human iPSC-derived cardiomyocytes with RBPMS gene deletion have a similar blockade to cytokinesis. Sequencing analysis revealed that RBPMS plays a role in RNA splicing and influences RNAs involved in cytoskeletal signaling pathways. We found that RBPMS mediates the isoform switching of the heart-enriched LIM domain protein Pdlim5. The loss of Rbpms leads to an abnormal accumulation of Pdlim5-short isoforms, disrupting cardiomyocyte cytokinesis. Our findings connect premature cardiomyocyte binucleation to noncompaction cardiomyopathy and highlight the role of RBPMS in this process.
Collapse
Affiliation(s)
- Peiheng Gan
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhaoning Wang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cellular and Molecular Medicine, University of California, San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Maria Gabriela Morales
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yu Zhang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ning Liu
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
8
|
Soetkamp D, Gallet R, Parker SJ, Holewinski R, Venkatraman V, Peck K, Goldhaber JI, Marbán E, Van Eyk JE. Myofilament Phosphorylation in Stem Cell Treated Diastolic Heart Failure. Circ Res 2021; 129:1125-1140. [PMID: 34641704 DOI: 10.1161/circresaha.119.316311] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
RATIONALE Phosphorylation of sarcomeric proteins has been implicated in heart failure with preserved ejection fraction (HFpEF); such changes may contribute to diastolic dysfunction by altering contractility, cardiac stiffness, Ca2+-sensitivity, and mechanosensing. Treatment with cardiosphere-derived cells (CDCs) restores normal diastolic function, attenuates fibrosis and inflammation, and improves survival in a rat HFpEF model. OBJECTIVE Phosphorylation changes that underlie HFpEF and those reversed by CDC therapy, with a focus on the sarcomeric subproteome were analyzed. METHODS AND RESULTS Dahl salt-sensitive rats fed a high-salt diet, with echocardiographically verified diastolic dysfunction, were randomly assigned to either intracoronary CDCs or placebo. Dahl salt-sensitive rats receiving low salt diet served as controls. Protein and phosphorylated Ser, Thr, and Tyr residues from left ventricular tissue were quantified by mass spectrometry. HFpEF hearts exhibited extensive hyperphosphorylation with 98% of the 529 significantly changed phospho-sites increased compared with control. Of those, 39% were located within the sarcomeric subproteome, with a large group of proteins located or associated with the Z-disk. CDC treatment partially reverted the hyperphosphorylation, with 85% of the significantly altered 76 residues hypophosphorylated. Bioinformatic upstream analysis of the differentially phosphorylated protein residues revealed PKC as the dominant putative regulatory kinase. PKC isoform analysis indicated increases in PKC α, β, and δ concentration, whereas CDC treatment led to a reversion of PKCβ. Use of PKC isoform specific inhibition and overexpression of various PKC isoforms strongly suggests that PKCβ is the dominant kinase involved in hyperphosphorylation in HFpEF and is altered with CDC treatment. CONCLUSIONS Increased protein phosphorylation at the Z-disk is associated with diastolic dysfunction, with PKC isoforms driving most quantified phosphorylation changes. Because CDCs reverse the key abnormalities in HFpEF and selectively reverse PKCβ upregulation, PKCβ merits being classified as a potential therapeutic target in HFpEF, a disease notoriously refractory to medical intervention.
Collapse
Affiliation(s)
- Daniel Soetkamp
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Romain Gallet
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Sarah J Parker
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | | | - Kiel Peck
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | |
Collapse
|
9
|
Huang X, Qu R, Peng Y, Yang Y, Fan T, Sun B, Khan AU, Wu S, Wei K, Xu C, Dai J, Ouyang J, Zhong S. Mechanical Sensing Element PDLIM5 Promotes Osteogenesis of Human Fibroblasts by Affecting the Activity of Microfilaments. Biomolecules 2021; 11:biom11050759. [PMID: 34069539 PMCID: PMC8161207 DOI: 10.3390/biom11050759] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 12/23/2022] Open
Abstract
Human skin fibroblasts (HSFs) approximate the multidirectional differentiation potential of mesenchymal stem cells, so they are often used in differentiation, cell cultures, and injury repair. They are an important seed source in the field of bone tissue engineering. However, there are a few studies describing the mechanism of osteogenic differentiation of HSFs. Here, osteogenic induction medium was used to induce fibroblasts to differentiate into osteoblasts, and the role of the mechanical sensitive element PDLIM5 in microfilament-mediated osteogenic differentiation of human fibroblasts was evaluated. The depolymerization of microfilaments inhibited the expression of osteogenesis-related proteins and alkaline phosphatase activity of HSFs, while the polymerization of microfilaments enhanced the osteogenic differentiation of HSFs. The evaluation of potential protein molecules affecting changes in microfilaments showed that during the osteogenic differentiation of HSFs, the expression of PDLIM5 increased with increasing induction time, and decreased under the state of microfilament depolymerization. Lentivirus-mediated PDLIM5 knockdown by shRNA weakened the osteogenic differentiation ability of HSFs and inhibited the expression and morphological changes of microfilament protein. The inhibitory effect of knocking down PDLIM5 on HSF osteogenic differentiation was reversed by a microfilament stabilizer. Taken together, these data suggest that PDLIM5 can mediate the osteogenic differentiation of fibroblasts by affecting the formation and polymerization of microfilaments.
Collapse
Affiliation(s)
- Xiaolan Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Yan Peng
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Yuchao Yang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Tingyu Fan
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Bing Sun
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Shutong Wu
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
| | - Kuanhai Wei
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China;
| | - Chujiang Xu
- Department of Orthopedics, TCM-Integrated Hospital, Southern Medical University, Guangzhou 510000, China;
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
- Correspondence: (J.D.); (J.O.); (S.Z.); Tel.: +86-(20)-6164-8842 (J.D.); +86-(20)-6164-8199 (J.O.); +86-(20)-6164-8200 (S.Z.)
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
- Correspondence: (J.D.); (J.O.); (S.Z.); Tel.: +86-(20)-6164-8842 (J.D.); +86-(20)-6164-8199 (J.O.); +86-(20)-6164-8200 (S.Z.)
| | - Shizhen Zhong
- Guangdong Provincial Key Laboratory of Medical Biomechanics & Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China; (X.H.); (R.Q.); (Y.P.); (Y.Y.); (T.F.); (B.S.); (A.U.K.); (S.W.)
- Correspondence: (J.D.); (J.O.); (S.Z.); Tel.: +86-(20)-6164-8842 (J.D.); +86-(20)-6164-8199 (J.O.); +86-(20)-6164-8200 (S.Z.)
| |
Collapse
|
10
|
Huang X, Qu R, Ouyang J, Zhong S, Dai J. An Overview of the Cytoskeleton-Associated Role of PDLIM5. Front Physiol 2020; 11:975. [PMID: 32848888 PMCID: PMC7426503 DOI: 10.3389/fphys.2020.00975] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/16/2020] [Indexed: 01/08/2023] Open
Abstract
Regenerative medicine represented by stem cell technology has become one of the pillar medical technologies for human disease treatment. Cytoskeleton plays important roles in maintaining cell morphology, bearing external forces, and maintaining the effectiveness of cell internal structure, among which cytoskeleton related proteins are involved in and play an indispensable role in the changes of cytoskeleton. PDLIM5 is a cytoskeleton-related protein that, like other cytoskeletal proteins, acts as a binding protein. PDZ and LIM domain 5 (PDLIM5), also known as ENH (Enigma homolog), is a cytoplasmic protein with a molecular mass of about 63 KDa that consists of a PDZ domain at the N-terminus and three LIM domains at the C-terminus. PDLIM5 binds to the cytoskeleton and membrane proteins through its PDZ domain and interacts with various signaling molecules, including protein kinases and transcription factors, through its LIM domain. As a cytoskeleton-related protein, PDLIM5 plays an important role in regulating cell proliferation, differentiation and cell fate decision in multiple tissues and cell types. In this review, we briefly summarize the state of knowledge on the PDLIM5 gene, structural properties, and molecular functional mechanisms of the PDLIM5 protein, and its role in cells, tissues, and organ systems, and describe the possible underlying molecular signaling pathways. In the last part of this review, we will focus on discussing the limitations of existing research and the future prospects of PDLIM5 research in turn.
Collapse
Affiliation(s)
- Xiaolan Huang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shizhen Zhong
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Tanaka R, Miyata S, Yamaguchi M, Yoshida H. Role of the smallish gene during Drosophila eye development. Gene 2019; 684:10-19. [DOI: 10.1016/j.gene.2018.10.056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/15/2018] [Accepted: 10/19/2018] [Indexed: 02/06/2023]
|
12
|
Gregorich ZR, Patel JR, Cai W, Lin Z, Heurer R, Fitzsimons DP, Moss RL, Ge Y. Deletion of Enigma Homologue from the Z-disc slows tension development kinetics in mouse myocardium. J Gen Physiol 2019; 151:670-679. [PMID: 30642915 PMCID: PMC6504290 DOI: 10.1085/jgp.201812214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022] Open
Abstract
Enigma Homologue (ENH) is a component of the Z-disc, a structure that anchors actin filaments in the contractile unit of muscle, the sarcomere. Cardiac-specific ablation of ENH protein expression causes contractile dysfunction that ultimately culminates in dilated cardiomyopathy. However, whether ENH is involved in the regulation of myocardial contractility is unknown. To determine if ENH is required for the mechanical activity of cardiac muscle, we analyze muscle mechanics of isolated trabeculae from the hearts of ENH +/+ and ENH -/- mice. We detected no differences in steady-state mechanical properties but show that when muscle fibers are allowed to relax and then are restretched, the rate at which tension redevelops is depressed in ENH -/- mouse myocardium relative to that in ENH +/+ myocardium. SDS-PAGE analysis demonstrated that the expression of β-myosin heavy chain is increased in ENH -/- mouse myocardium, which could partially, but not completely, account for the depression in tension redevelopment kinetics. Using top-down proteomics analysis, we found that the expression of other thin/thick filament regulatory proteins is unaltered, although the phosphorylation of a cardiac troponin T isoform, cardiac troponin I, and myosin regulatory light chain is decreased in ENH -/- mouse myocardium. Nevertheless, these alterations are very small and thus insufficient to explain slowed tension redevelopment kinetics in ENH -/- mouse myocardium. These data suggest that the ENH protein influences tension redevelopment kinetics in mouse myocardium, possibly by affecting cross-bridge cycling kinetics. Previous studies also indicate that ablation of specific Z-disc proteins in myocardium slows contraction kinetics, which could also be a contributing factor in this study.
Collapse
Affiliation(s)
- Zachery R Gregorich
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI.,Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI
| | - Jitandrakumar R Patel
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI.,University of Wisconsin-Madison Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI
| | - Wenxuan Cai
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI.,Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI
| | - Ziqing Lin
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI.,Human Proteomics Program, University of Wisconsin-Madison, Madison, WI
| | - Rachel Heurer
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI
| | - Daniel P Fitzsimons
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI
| | - Richard L Moss
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI .,University of Wisconsin-Madison Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI.,Human Proteomics Program, University of Wisconsin-Madison, Madison, WI
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI .,Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI.,University of Wisconsin-Madison Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI.,Human Proteomics Program, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
13
|
Sahebekhtiari N, Fernandez-Guerra P, Nochi Z, Carlsen J, Bross P, Palmfeldt J. Deficiency of the mitochondrial sulfide regulator ETHE1 disturbs cell growth, glutathione level and causes proteome alterations outside mitochondria. Biochim Biophys Acta Mol Basis Dis 2018; 1865:126-135. [PMID: 30391543 DOI: 10.1016/j.bbadis.2018.10.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/14/2018] [Accepted: 10/30/2018] [Indexed: 01/15/2023]
Abstract
The mitochondrial enzyme ETHE1 is a persulfide dioxygenase essential for cellular sulfide detoxification, and its deficiency causes the severe and complex inherited metabolic disorder ethylmalonic encephalopathy (EE). In spite of well-described clinical symptoms of the disease, detailed cellular and molecular characterization is still ambiguous. Cellular redox regulation has been described to be influenced in ETHE1 deficient cells, and to clarify this further we applied image cytometry and detected decreased levels of reduced glutathione (GSH) in cultivated EE patient fibroblast cells. Cell growth initiation of the EE patient cells was impaired, whereas cell cycle regulation was not. Furthermore, Seahorse metabolic analyzes revealed decreased extracellular acidification, i. e. decreased lactate formation from glycolysis, in the EE patient cells. TMT-based large-scale proteomics was subsequently performed to broadly elucidate cellular consequences of the ETHE1 deficiency. More than 130 proteins were differentially regulated, of which the majority were non-mitochondrial. The proteomics data revealed a link between ETHE1-deficiency and down-regulation of several ribosomal proteins and LIM domain proteins important for cellular maintenance, and up-regulation of cell surface glycoproteins. Furthermore, several proteins of endoplasmic reticulum (ER) were perturbed including proteins influencing disulfide bond formation (e.g. protein disulfide isomerases and peroxiredoxin 4) and calcium-regulated proteins. The results indicate that decreased level of reduced GSH and alterations in proteins of ribosomes, ER and of cell adhesion lie behind the disrupted cell growth of the EE patient cells.
Collapse
Affiliation(s)
- Navid Sahebekhtiari
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark
| | - Paula Fernandez-Guerra
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark
| | - Zahra Nochi
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark
| | - Jasper Carlsen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark
| | - Peter Bross
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University and Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200 Aarhus N, Denmark.
| |
Collapse
|
14
|
Konze SA, Werneburg S, Oberbeck A, Olmer R, Kempf H, Jara-Avaca M, Pich A, Zweigerdt R, Buettner FFR. Proteomic Analysis of Human Pluripotent Stem Cell Cardiomyogenesis Revealed Altered Expression of Metabolic Enzymes and PDLIM5 Isoforms. J Proteome Res 2017; 16:1133-1149. [DOI: 10.1021/acs.jproteome.6b00534] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Sarah A. Konze
- Institute
of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
- REBIRTH
Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| | - Sebastian Werneburg
- Institute
of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
- REBIRTH
Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| | - Astrid Oberbeck
- Institute
of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
- REBIRTH
Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| | - Ruth Olmer
- Leibniz
Research Laboratories for Biotechnology and Artificial Organs, Department
of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
- REBIRTH
Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| | - Henning Kempf
- Leibniz
Research Laboratories for Biotechnology and Artificial Organs, Department
of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
- REBIRTH
Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| | - Monica Jara-Avaca
- Leibniz
Research Laboratories for Biotechnology and Artificial Organs, Department
of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
- REBIRTH
Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas Pich
- Institute
of Toxicology, Core Facility Proteomics, Hannover Medical School, 30625 Hannover, Germany
| | - Robert Zweigerdt
- Leibniz
Research Laboratories for Biotechnology and Artificial Organs, Department
of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany
- REBIRTH
Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| | - Falk F. R. Buettner
- Institute
of Clinical Biochemistry, Hannover Medical School, 30625 Hannover, Germany
- REBIRTH
Cluster of Excellence, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
15
|
Reimann L, Wiese H, Leber Y, Schwäble AN, Fricke AL, Rohland A, Knapp B, Peikert CD, Drepper F, van der Ven PFM, Radziwill G, Fürst DO, Warscheid B. Myofibrillar Z-discs Are a Protein Phosphorylation Hot Spot with Protein Kinase C (PKCα) Modulating Protein Dynamics. Mol Cell Proteomics 2016; 16:346-367. [PMID: 28028127 DOI: 10.1074/mcp.m116.065425] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Indexed: 11/06/2022] Open
Abstract
The Z-disc is a protein-rich structure critically important for the development and integrity of myofibrils, which are the contractile organelles of cross-striated muscle cells. We here used mouse C2C12 myoblast, which were differentiated into myotubes, followed by electrical pulse stimulation (EPS) to generate contracting myotubes comprising mature Z-discs. Using a quantitative proteomics approach, we found significant changes in the relative abundance of 387 proteins in myoblasts versus differentiated myotubes, reflecting the drastic phenotypic conversion of these cells during myogenesis. Interestingly, EPS of differentiated myotubes to induce Z-disc assembly and maturation resulted in increased levels of proteins involved in ATP synthesis, presumably to fulfill the higher energy demand of contracting myotubes. Because an important role of the Z-disc for signal integration and transduction was recently suggested, its precise phosphorylation landscape further warranted in-depth analysis. We therefore established, by global phosphoproteomics of EPS-treated contracting myotubes, a comprehensive site-resolved protein phosphorylation map of the Z-disc and found that it is a phosphorylation hotspot in skeletal myocytes, underscoring its functions in signaling and disease-related processes. In an illustrative fashion, we analyzed the actin-binding multiadaptor protein filamin C (FLNc), which is essential for Z-disc assembly and maintenance, and found that PKCα phosphorylation at distinct serine residues in its hinge 2 region prevents its cleavage at an adjacent tyrosine residue by calpain 1. Fluorescence recovery after photobleaching experiments indicated that this phosphorylation modulates FLNc dynamics. Moreover, FLNc lacking the cleaved Ig-like domain 24 exhibited remarkably fast kinetics and exceedingly high mobility. Our data set provides research community resource for further identification of kinase-mediated changes in myofibrillar protein interactions, kinetics, and mobility that will greatly advance our understanding of Z-disc dynamics and signaling.
Collapse
Affiliation(s)
- Lena Reimann
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Heike Wiese
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Yvonne Leber
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Anja N Schwäble
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anna L Fricke
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Anne Rohland
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Bettina Knapp
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Christian D Peikert
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Friedel Drepper
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Peter F M van der Ven
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Gerald Radziwill
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,§BIOSS Centre for Biological Signalling Studies, University of Freiburg
| | - Dieter O Fürst
- ¶Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Bettina Warscheid
- From the ‡Department of Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; .,§BIOSS Centre for Biological Signalling Studies, University of Freiburg
| |
Collapse
|
16
|
Ito J, Iijima M, Yoshimoto N, Niimi T, Kuroda S, Maturana AD. RBM20 and RBM24 cooperatively promote the expression of short enh splice variants. FEBS Lett 2016; 590:2262-74. [PMID: 27289039 DOI: 10.1002/1873-3468.12251] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 04/29/2016] [Accepted: 05/19/2016] [Indexed: 12/31/2022]
Abstract
PDZ-LIM protein ENH1 is a scaffold protein for protein kinases and transcriptional regulators. While ENH1 promotes the hypertrophic growth of cardiomyocytes, its short splice variant (ENH3) prevents the hypertrophic growth. The mechanism underlying the alternative splicing of enh mRNA between ENH short and long isoforms has remained unknown. Here, we found that two splicing factors, RNA-binding motif 20 (RBM20) and RNA-binding motif 24 (RBM24) together promoted the expression of short enh splice variants and bound the 5' intronic region of exon 11 containing an in-phase stop codon. In addition, expression of both RBMs is repressed by hypertrophic stimulations. Collectively, our results suggest that, in healthy conditions, RBM20 and RBM24 cooperate to promote the expression of short ENH isoforms.
Collapse
Affiliation(s)
- Jumpei Ito
- Graduate School of Bioagricultural Sciences, Nagoya University, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Masumi Iijima
- The Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Osaka, Japan
| | - Nobuo Yoshimoto
- The Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Osaka, Japan
| | - Tomoaki Niimi
- Graduate School of Bioagricultural Sciences, Nagoya University, Japan
| | - Shun'ichi Kuroda
- Graduate School of Bioagricultural Sciences, Nagoya University, Japan.,The Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Osaka, Japan
| | - Andrés D Maturana
- Graduate School of Bioagricultural Sciences, Nagoya University, Japan
| |
Collapse
|
17
|
MicroRNA-17-92 regulates myoblast proliferation and differentiation by targeting the ENH1/Id1 signaling axis. Cell Death Differ 2016; 23:1658-69. [PMID: 27315298 PMCID: PMC5041193 DOI: 10.1038/cdd.2016.56] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 05/19/2015] [Accepted: 05/20/2016] [Indexed: 12/21/2022] Open
Abstract
Myogenesis is an important biological process that occurs during both skeletal muscle regeneration and postnatal growth. Growing evidence points to the critical role of microRNAs (miRNAs) in myogenesis. Our analysis of miRNA expression patterns reveal that miRNAs of miR-17-92 cluster are dramatically downregulated in C2C12 cells after myogenesis stimulation, are strongly induced in mouse skeletal muscle after injury and decrease steadily thereafter and are downregulated with age in skeletal muscle during mouse and porcine postnatal growth. However, their roles in muscle developmental processes remain elusive. We show that the miR-17-92 cluster promotes mouse myoblast proliferation but inhibits myotube formation. miR-17, -20a and -92a target the actin-associated protein enigma homolog 1 (ENH1). The silencing of ENH1 increased the nuclear accumulation of the inhibitor of differentiation 1 (Id1) and represses myogenic differentiation. Furthermore, the injection of adenovirus expressing miR-20a into the tibialia anterior muscle downregulates ENH1 and delays regeneration. In addition, the downregulation of miR-17-92 during myogenesis is transcriptionally regulated by E2F1. Overall, our results reveal a E2F1/miR-17-92/ENH1/Id1 regulatory axis during myogenesis.
Collapse
|
18
|
Nakatani M, Ito J, Koyama R, Iijima M, Yoshimoto N, Niimi T, Kuroda S, Maturana AD. Scaffold protein enigma homolog 1 overcomes the repression of myogenesis activation by inhibitor of DNA binding 2. Biochem Biophys Res Commun 2016; 474:413-420. [PMID: 27114303 DOI: 10.1016/j.bbrc.2016.04.119] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 04/22/2016] [Indexed: 01/30/2023]
Abstract
Enigma Homolog 1 (ENH1) is a scaffold protein for signaling proteins and transcription factors. Previously, we reported that ENH1 overexpression promotes the differentiation of C2C12 myoblasts. However, the molecular mechanism underlying the role of ENH1 in the C2C12 cells differentiation remains elusive. ENH1 was shown to inhibit the proliferation of neuroblastoma cells by sequestering Inhibitor of DNA binding protein 2 (Id2) in the cytosol. Id2 is a repressor of basic Helix-Loop-Helix transcription factors activity and prevents myogenesis. Here, we found that ENH1 overcome the Id2 repression of C2C12 cells myogenic differentiation and that ENH1 overexpression promotes mice satellite cells activation, the first step toward myogenic differentiation. In addition, we show that ENH1 interacted with Id2 in C2C12 cells and mice satellite cells. Collectively, our results suggest that ENH1 plays an important role in the activation of myogenesis through the repression of Id2 activity.
Collapse
Affiliation(s)
- Miyuki Nakatani
- Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Nagoya, 464-8106, Japan
| | - Jumpei Ito
- Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Nagoya, 464-8106, Japan; Japan Society for the Promotion of Science, Tokyo, 102-0083, Japan
| | - Riko Koyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Nagoya, 464-8106, Japan
| | - Masumi Iijima
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Nobuo Yoshimoto
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Tomoaki Niimi
- Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Nagoya, 464-8106, Japan
| | - Shun'ichi Kuroda
- Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Nagoya, 464-8106, Japan; The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka, 567-0047, Japan
| | - Andrés D Maturana
- Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Nagoya, 464-8106, Japan.
| |
Collapse
|
19
|
Cheng H, Chen T, Tor M, Park D, Zhou Q, Huang JB, Khatib N, Rong L, Zhou G. A High-Throughput Screening Platform Targeting PDLIM5 for Pulmonary Hypertension. ACTA ACUST UNITED AC 2016; 21:333-41. [DOI: 10.1177/1087057115625924] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/16/2015] [Indexed: 11/16/2022]
Abstract
Pulmonary arterial hypertension is a complex disease with multiple etiologic factors. PDLIM5, a member of the Enigma subfamily of PDZ and LIM domain protein family, contains an N-terminal PDZ domain and three LIM domains at its C-terminus. We have previously shown that overexpression of PDLIM5 prevents hypoxia-induced pulmonary hypertension (PH), and deletion of PDLIM5 in smooth muscle cells enhances hypoxia-induced PH in vivo. These results suggest that PDLIM5 may be a novel therapeutic target of PH. In this study, we aim to establish a high-throughput screening platform for PDLIM5-targeted drug discovery. We generated a stable mink lung epithelial cell line (MLEC) containing a transforming growth factor–β/Smad luciferase reporter with lentivirus-mediated suppression of PDLIM5 (MLEC-shPDLIM5) and measured levels of Smad2/3 and pSmad2/3. We found that in MLEC, suppression of PDLIM5 decreased Smad-dependent luciferase activity, Smad3, and pSmad3. We used MLEC-shPDLIM5 and a control cell line (MLEC-shCTL) to screen the Prestwick library (1200 compounds) and identified and validated paclitaxel as a PDLIM5 inhibitor in MLEC. Furthermore, we showed that paclitaxel inhibited Smad2 expression and Smad3 phosphorylation in A549 cells. Our study suggests that this system is robust and suitable for PDLIM5-targeted drug discovery.
Collapse
Affiliation(s)
- Han Cheng
- Department of Microbiology and Immunology, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Tianji Chen
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Merve Tor
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Deborah Park
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Qiyuan Zhou
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Jason B. Huang
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Nour Khatib
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
| | - Guofei Zhou
- Department of Pediatrics, College of Medicine University of Illinois at Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| |
Collapse
|
20
|
PDLIM1 inhibits NF-κB-mediated inflammatory signaling by sequestering the p65 subunit of NF-κB in the cytoplasm. Sci Rep 2015; 5:18327. [PMID: 26679095 PMCID: PMC4683373 DOI: 10.1038/srep18327] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/16/2015] [Indexed: 12/03/2022] Open
Abstract
Understanding the regulatory mechanisms for the NF-κB transcription factor is key to control inflammation. IκBα maintains NF-κB in an inactive form in the cytoplasm of unstimulated cells, whereas nuclear NF-κB in activated cells is degraded by PDLIM2, a nuclear ubiquitin E3 ligase that belongs to a LIM protein family. How NF-κB activation is negatively controlled, however, is not completely understood. Here we show that PDLIM1, another member of LIM proteins, negatively regulates NF-κB-mediated signaling in the cytoplasm. PDLIM1 sequestered p65 subunit of NF-κB in the cytoplasm and suppressed its nuclear translocation in an IκBα-independent, but α-actinin-4-dependent manner. Consistently, PDLIM1 deficiency lead to increased levels of nuclear p65 protein, and thus enhanced proinflammatory cytokine production in response to innate stimuli. These studies reveal an essential role of PDLIM1 in suppressing NF-κB activation and suggest that LIM proteins comprise a new family of negative regulators of NF-κB signaling through different mechanisms.
Collapse
|
21
|
PDLIM7 is a novel target of the ubiquitin ligase Nedd4-1 in skeletal muscle. Biochem J 2015; 473:267-76. [PMID: 26556890 DOI: 10.1042/bj20150222] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 11/10/2015] [Indexed: 01/07/2023]
Abstract
Skeletal muscle atrophy remains a complication occurring both as a natural response to muscle disuse and as a pathophysiological response to illness such as diabetes mellitus and nerve injury, such as traumatic muscle denervation. The ubiquitin-proteasome system (UPS) is the predominant proteolytic machinery responsible for atrophy of skeletal muscle, and Nedd4-1 (neural precursor cell-expressed developmentally down-regulated 4-1) is one of a series of E3 ubiquitin ligases identified to mediate inactivity-induced muscle wasting. Targets of Nedd4-1 mediated ubiquitination in skeletal muscle remain poorly understood. In the present study, we identified PDLIM7 (PDZ and LIM domain 7, Enigma), a member of the PDZ-LIM family of proteins, as a novel target of Nedd4-1 in skeletal muscle. The PDZ-LIM family of proteins is known to regulate muscle development and function. We show that Nedd4-1 expression in muscle atrophied by denervation is co-incident with a decrease in PDLIM7 and that PDLIM7 protein levels are stabilized in denervated muscle of Nedd4-1 skeletal muscle-specific knockout mice (SMS-KO). Exogenous PDLIM7 and Nedd4-1 transfected into human embryonic kidney (HEK)293 cells co-immunoprecipitate through binding between the PY motif of PDLIM7 and the second and third WW domains of Nedd4-1 and endogenous PDLIM7 and Nedd4-1 interact in the cytoplasm of differentiated C2C12 myotubes, leading to PDLIM7 ubiquitination. These results identify PDLIM7 as a bona fide skeletal muscle substrate of Nedd4-1 and suggest that this interaction may underlie the progression of skeletal muscle atrophy. This offers a novel therapeutic target that could be potentially used to attenuate muscle atrophy.
Collapse
|
22
|
Scaffold protein enigma homolog activates CREB whereas a short splice variant prevents CREB activation in cardiomyocytes. Cell Signal 2015; 27:2425-33. [PMID: 26365342 DOI: 10.1016/j.cellsig.2015.09.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 09/09/2015] [Indexed: 01/27/2023]
Abstract
Enigma Homolog (ENH1 or Pdlim5) is a scaffold protein composed of an N-terminal PDZ domain and three LIM domains at the C-terminal end. The enh gene encodes for several splice variants with opposing functions. ENH1 promotes cardiomyocytes hypertrophy whereas ENH splice variants lacking LIM domains prevent it. ENH1 interacts with various Protein Kinase C (PKC) isozymes and Protein Kinase D1 (PKD1). In addition, the binding of ENH1's LIM domains to PKC is sufficient to activate the kinase without stimulation. The downstream events of the ENH1-PKC/PKD1 complex remain unknown. PKC and PKD1 are known to phosphorylate the transcription factor cAMP-response element binding protein (CREB). We tested whether ENH1 could play a role in the activation of CREB. We found that, in neonatal rat ventricular cardiomyocytes, ENH1 interacts with CREB, is necessary for the phosphorylation of CREB at ser133, and the activation of CREB-dependent transcription. On the contrary, the overexpression of ENH3, a LIM-less splice variant, inhibited the phosphorylation of CREB. ENH3 overexpression or shRNA knockdown of ENH1 prevented the CREB-dependent transcription. Our results thus suggest that ENH1 plays an essential role in CREB's activation and dependent transcription in cardiomyocytes. At the opposite, ENH3 prevents the CREB transcriptional activity. In conclusion, these results provide a first molecular explanation to the opposing functions of ENH splice variants.
Collapse
|
23
|
Ahn BY, Saldanha-Gama RFG, Rahn JJ, Hao X, Zhang J, Dang NH, Alshehri M, Robbins SM, Senger DL. Glioma invasion mediated by the p75 neurotrophin receptor (p75(NTR)/CD271) requires regulated interaction with PDLIM1. Oncogene 2015; 35:1411-22. [PMID: 26119933 PMCID: PMC4800290 DOI: 10.1038/onc.2015.199] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 04/23/2015] [Accepted: 05/10/2015] [Indexed: 01/05/2023]
Abstract
The invasive nature of glioblastoma renders them incurable by current therapeutic interventions. Using a novel invasive human glioma model, we previously identified the neurotrophin receptor p75NTR (aka CD271) as a mediator of glioma invasion. Herein, we provide evidence that preventing phosphorylation of p75NTR on S303 by pharmacological inhibition of PKA, or by a mutational strategy (S303G), cripples p75NTR-mediated glioma invasion resulting in serine phosphorylation within the C-terminal PDZ-binding motif (SPV) of p75NTR. Consistent with this, deletion (ΔSPV) or mutation (SPM) of the PDZ motif results in abrogation of p75NTR-mediated invasion. Using a peptide-based strategy, we identified PDLIM1 as a novel signaling adaptor for p75NTR and provide the first evidence for a regulated interaction via S425 phosphorylation. Importantly, PDLIM1 was shown to interact with p75NTR in highly invasive patient-derived glioma stem cells/tumor-initiating cells and shRNA knockdown of PDLIM1 in vitro and in vivo results in complete ablation of p75NTR-mediated invasion. Collectively, these data demonstrate a requirement for a regulated interaction of p75NTR with PDLIM1 and suggest that targeting either the PDZ domain interactions and/or the phosphorylation of p75NTR by PKA could provide therapeutic strategies for patients with glioblastoma.
Collapse
Affiliation(s)
- B Y Ahn
- Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada.,Hughes Childhood Cancer Program, University of Calgary, Calgary, Alberta, Canada.,Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - R F G Saldanha-Gama
- Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Oncology, University of Calgary, Calgary, Alberta, Canada.,Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
| | - J J Rahn
- Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada.,Hughes Childhood Cancer Program, University of Calgary, Calgary, Alberta, Canada.,Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - X Hao
- Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada.,Hughes Childhood Cancer Program, University of Calgary, Calgary, Alberta, Canada.,Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - J Zhang
- Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada.,Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
| | - N-H Dang
- Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Oncology, University of Calgary, Calgary, Alberta, Canada.,Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
| | - M Alshehri
- Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Oncology, University of Calgary, Calgary, Alberta, Canada.,Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
| | - S M Robbins
- Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada.,Hughes Childhood Cancer Program, University of Calgary, Calgary, Alberta, Canada.,Department of Oncology, University of Calgary, Calgary, Alberta, Canada.,Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
| | - D L Senger
- Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada.,Hughes Childhood Cancer Program, University of Calgary, Calgary, Alberta, Canada.,Department of Oncology, University of Calgary, Calgary, Alberta, Canada.,Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
24
|
Chen T, Zhou G, Zhou Q, Tang H, Ibe JCF, Cheng H, Gou D, Chen J, Yuan JXJ, Raj JU. Loss of microRNA-17∼92 in smooth muscle cells attenuates experimental pulmonary hypertension via induction of PDZ and LIM domain 5. Am J Respir Crit Care Med 2015; 191:678-92. [PMID: 25647182 DOI: 10.1164/rccm.201405-0941oc] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
RATIONALE Recent studies suggest that microRNAs (miRNAs) play important roles in regulation of pulmonary artery smooth muscle cell (PASMC) phenotype and are implicated in pulmonary arterial hypertension (PAH). However, the underlying molecular mechanisms remain elusive. OBJECTIVES This study aims to understand the mechanisms regulating PASMC proliferation and differentiation by microRNA-17∼92 (miR-17∼92) and to elucidate its implication in PAH. METHODS We generated smooth muscle cell (SMC)-specific miR-17∼92 and PDZ and LIM domain 5 (PDLIM5) knockout mice and overexpressed miR-17∼92 and PDLIM5 by injection of miR-17∼92 mimics or PDLIM5-V5-His plasmids and measured their responses to hypoxia. We used miR-17∼92 mimics, inhibitors, overexpression vectors, small interfering RNAs against PDLIM5, Smad, and transforming growth factor (TGF)-β to determine the role of miR-17∼92 and its downstream targets in PASMC proliferation and differentiation. MEASUREMENTS AND MAIN RESULTS We found that human PASMC (HPASMC) from patients with PAH expressed decreased levels of the miR-17∼92 cluster, TGF-β, and SMC markers. Overexpression of miR-17∼92 increased and restored the expression of TGF-β3, Smad3, and SMC markers in HPASMC of normal subjects and patients with idiopathic PAH, respectively. Knockdown of Smad3 but not Smad2 prevented miR-17∼92-induced expression of SMC markers. SMC-specific knockout of miR-17∼92 attenuated hypoxia-induced pulmonary hypertension (PH) in mice, whereas reconstitution of miR-17∼92 restored hypoxia-induced PH in these mice. We also found that PDLIM5 is a direct target of miR-17/20a, and hypertensive HPASMC and mouse PASMC expressed elevated PDLIM5 levels. Suppression of PDLIM5 increased expression of SMC markers and enhanced TGF-β/Smad2/3 activity in vitro and enhanced hypoxia-induced PH in vivo, whereas overexpression of PDLIM5 attenuated hypoxia-induced PH. CONCLUSIONS We provided the first evidence that miR-17∼92 inhibits PDLIM5 to induce the TGF-β3/SMAD3 pathway, contributing to the pathogenesis of PAH.
Collapse
|
25
|
Mu Y, Jing R, Peter AK, Lange S, Lin L, Zhang J, Ouyang K, Fang X, Veevers J, Zhou X, Evans SM, Cheng H, Chen J. Cypher and Enigma homolog protein are essential for cardiac development and embryonic survival. J Am Heart Assoc 2015; 4:jah3966. [PMID: 25944877 PMCID: PMC4599425 DOI: 10.1161/jaha.115.001950] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background The striated muscle Z-line, a multiprotein complex at the boundary between sarcomeres, plays an integral role in maintaining striated muscle structure and function. Multiple Z-line-associated proteins have been identified and shown to play an increasingly important role in the pathogenesis of human cardiomyopathy. Cypher and its close homologue, Enigma homolog protein (ENH), are 2 Z-line proteins previously shown to be individually essential for maintenance of postnatal cardiac function and stability of the Z-line during muscle contraction, but dispensable for cardiac myofibrillogenesis and development. Methods and Results The current studies were designed to test whether Cypher and ENH play redundant roles during embryonic development. Here, we demonstrated that mice lacking both ENH and Cypher exhibited embryonic lethality and growth retardation. Lethality in double knockout embryos was associated with cardiac dilation and abnormal Z-line structure. In addition, when ENH was ablated in conjunction with selective ablation of either Cypher short isoforms (CypherS), or Cypher long isoforms (CypherL), only the latter resulted in embryonic lethality. Conclusions Cypher and ENH redundantly play an essential role in sustaining Z-line structure from the earliest stages of cardiac function, and are redundantly required to maintain normal embryonic heart function and embryonic viability.
Collapse
Affiliation(s)
- Yongxin Mu
- Department of Medicine, University of California San Diego, La Jolla, CA (Y.M., R.J., A.K.P., S.L., L.L., J.Z., X.F., J.V., S.M.E., J.C.)
| | - Ran Jing
- Department of Medicine, University of California San Diego, La Jolla, CA (Y.M., R.J., A.K.P., S.L., L.L., J.Z., X.F., J.V., S.M.E., J.C.) Xiangya Hospital, Central South University, Changsha, China (R.J., X.Z.)
| | - Angela K Peter
- Department of Medicine, University of California San Diego, La Jolla, CA (Y.M., R.J., A.K.P., S.L., L.L., J.Z., X.F., J.V., S.M.E., J.C.)
| | - Stephan Lange
- Department of Medicine, University of California San Diego, La Jolla, CA (Y.M., R.J., A.K.P., S.L., L.L., J.Z., X.F., J.V., S.M.E., J.C.)
| | - Lizhu Lin
- Department of Medicine, University of California San Diego, La Jolla, CA (Y.M., R.J., A.K.P., S.L., L.L., J.Z., X.F., J.V., S.M.E., J.C.) Department of Medicine, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA (L.L., S.M.E.)
| | - Jianlin Zhang
- Department of Medicine, University of California San Diego, La Jolla, CA (Y.M., R.J., A.K.P., S.L., L.L., J.Z., X.F., J.V., S.M.E., J.C.)
| | - Kunfu Ouyang
- Key Laboratory of Chemical Genomics, Drug Discovery Center, Peking University Shenzhen Graduate School, Shenzhen, China (K.O.)
| | - Xi Fang
- Department of Medicine, University of California San Diego, La Jolla, CA (Y.M., R.J., A.K.P., S.L., L.L., J.Z., X.F., J.V., S.M.E., J.C.)
| | - Jennifer Veevers
- Department of Medicine, University of California San Diego, La Jolla, CA (Y.M., R.J., A.K.P., S.L., L.L., J.Z., X.F., J.V., S.M.E., J.C.)
| | - Xinmin Zhou
- Xiangya Hospital, Central South University, Changsha, China (R.J., X.Z.)
| | - Sylvia M Evans
- Department of Medicine, University of California San Diego, La Jolla, CA (Y.M., R.J., A.K.P., S.L., L.L., J.Z., X.F., J.V., S.M.E., J.C.) Department of Medicine, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA (L.L., S.M.E.)
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology, Program in Molecular Cell Biology, Zhejiang University School of Medicine, Hangzhou, China (H.C.)
| | - Ju Chen
- Department of Medicine, University of California San Diego, La Jolla, CA (Y.M., R.J., A.K.P., S.L., L.L., J.Z., X.F., J.V., S.M.E., J.C.)
| |
Collapse
|
26
|
Yan Y, Tsukamoto O, Nakano A, Kato H, Kioka H, Ito N, Higo S, Yamazaki S, Shintani Y, Matsuoka K, Liao Y, Asanuma H, Asakura M, Takafuji K, Minamino T, Asano Y, Kitakaze M, Takashima S. Augmented AMPK activity inhibits cell migration by phosphorylating the novel substrate Pdlim5. Nat Commun 2015; 6:6137. [PMID: 25635515 PMCID: PMC4317497 DOI: 10.1038/ncomms7137] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 12/12/2014] [Indexed: 12/12/2022] Open
Abstract
Augmented AMP-activated protein kinase (AMPK) activity inhibits cell migration, possibly contributing to the clinical benefits of chemical AMPK activators in preventing atherosclerosis, vascular remodelling and cancer metastasis. However, the underlying mechanisms remain largely unknown. Here we identify PDZ and LIM domain 5 (Pdlim5) as a novel AMPK substrate and show that it plays a critical role in the inhibition of cell migration. AMPK directly phosphorylates Pdlim5 at Ser177. Exogenous expression of phosphomimetic S177D-Pdlim5 inhibits cell migration and attenuates lamellipodia formation. Consistent with this observation, S177D-Pdlim5 suppresses Rac1 activity at the cell periphery and displaces the Arp2/3 complex from the leading edge. Notably, S177D-Pdlim5, but not WT-Pdlim5, attenuates the association with Rac1-specific guanine nucleotide exchange factors at the cell periphery. Taken together, our findings indicate that phosphorylation of Pdlim5 on Ser177 by AMPK mediates inhibition of cell migration by suppressing the Rac1-Arp2/3 signalling pathway. Augmented AMP-activated protein kinase (AMPK) activity inhibits cell migration through an unknown mechanism. Here, Yan et al. show that AMPK phosphorylates the novel substrate PDZ and LIM domain 5 (Pdlim5), and that phosphomimetic Pdlim5 impairs cell migration by disrupting the Rac1-Arp2/3 signalling pathway.
Collapse
Affiliation(s)
- Yi Yan
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Atsushi Nakano
- Depertment of Clinical Research and Development, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Hisakazu Kato
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hidetaka Kioka
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Noriaki Ito
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shuichiro Higo
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Satoru Yamazaki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Yasunori Shintani
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ken Matsuoka
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yulin Liao
- Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, 510515 Guangzhou, China
| | - Hiroshi Asanuma
- Depertment of Clinical Research and Development, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Masanori Asakura
- Depertment of Clinical Research and Development, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Kazuaki Takafuji
- Center for Research Education, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tetsuo Minamino
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masafumi Kitakaze
- Depertment of Clinical Research and Development, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Seiji Takashima
- 1] Department of Medical Biochemistry, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan [2] Japan Science and Technology Agency-Core Research for Evolutional Science and Technology (CREST), Kawaguchi 332-0012, Japan
| |
Collapse
|
27
|
Ren B, Li X, Zhang J, Fan J, Duan J, Chen Y. PDLIM5 mediates PKCε translocation in PMA-induced growth cone collapse. Cell Signal 2014; 27:424-35. [PMID: 25524223 DOI: 10.1016/j.cellsig.2014.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/28/2014] [Accepted: 12/10/2014] [Indexed: 11/16/2022]
Abstract
Growth cone collapse is a critical repulsive response to various guidance cues for axon guidance. Protein kinase C epsilon (PKCε) plays important regulation roles in such responses. Translocation of PKCε from cytoplasm to membrane is crucial to archive its regulatory roles in this process. We previously reported that PDLIM5 could selectively recruit PKCε to its specific substrate in neurons. However, the molecular mechanism of PKCε translocation in the neuronal growth cone collapse remains elusive. Here, we demonstrated that PDLIM5 and PKCε co-existed in the nerve growth cones. By interacting with α-actinin, but not β-actin or β-tubulin, PDLIM5 might contribute to regulation of remodeling of the microfilaments in neurons. Meanwhile, PDLIM5 could also bind to PKCε to form PDLIM5-PKCε complexes in growth cones. In the primary cultured neurons, activation of PKCε by PMA resulted in translocation of both PKCε and PDLIM5 from cytoplasm to the membrane. Knockdown of either PDLIM5 or PKCε rescued the neuron from PMA-induced growth cone collapse. Furthermore, in neurons, application of PDLIM5 shRNA or over-expression of PDLIM5 LIM1-3 mutants reduced the amount of PKCε in the membrane. Together, these results suggest that PDLIM5 acts as a scaffold protein by mediated PKCε translocated to the membrane in PMA-induced growth cone collapse.
Collapse
Affiliation(s)
- Bingyu Ren
- Neurobiology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiubo Li
- Neurobiology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jifeng Zhang
- Neurobiology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jinjin Fan
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jingjing Duan
- Neurobiology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuan Chen
- Neurobiology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
28
|
Bei Y, Hong P. A novel approach to minimize false discovery rate in genome-wide data analysis. BMC SYSTEMS BIOLOGY 2014; 7 Suppl 4:S1. [PMID: 24564975 PMCID: PMC3856609 DOI: 10.1186/1752-0509-7-s4-s1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND High-throughput technologies, such as DNA microarray, have significantly advanced biological and biomedical research by enabling researchers to carry out genome-wide screens. One critical task in analyzing genome-wide datasets is to control the false discovery rate (FDR) so that the proportion of false positive features among those called significant is restrained. Recently a number of FDR control methods have been proposed and widely practiced, such as the Benjamini-Hochberg approach, the Storey approach and Significant Analysis of Microarrays (SAM). METHODS This paper presents a straight-forward yet powerful FDR control method termed miFDR, which aims to minimize FDR when calling a fixed number of significant features. We theoretically proved that the strategy used by miFDR is able to find the optimal number of significant features when the desired FDR is fixed. RESULTS We compared miFDR with the BH approach, the Storey approach and SAM on both simulated datasets and public DNA microarray datasets. The results demonstrated that miFDR outperforms others by identifying more significant features under the same FDR cut-offs. Literature search showed that many genes called only by miFDR are indeed relevant to the underlying biology of interest. CONCLUSIONS FDR has been widely applied to analyzing high-throughput datasets allowed for rapid discoveries. Under the same FDR threshold, miFDR is capable to identify more significant features than its competitors at a compatible level of complexity. Therefore, it can potentially generate great impacts on biological and biomedical research. AVAILABILITY If interested, please contact the authors for getting miFDR.
Collapse
|
29
|
Novel targets of sulforaphane in primary cardiomyocytes identified by proteomic analysis. PLoS One 2013; 8:e83283. [PMID: 24349480 PMCID: PMC3859650 DOI: 10.1371/journal.pone.0083283] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 11/11/2013] [Indexed: 12/19/2022] Open
Abstract
Cardiovascular diseases represent the main cause of mortality in the industrialized world and the identification of effective preventive strategies is of fundamental importance. Sulforaphane, an isothiocyanate from cruciferous vegetables, has been shown to up-regulate phase II enzymes in cardiomyocytes and counteract oxidative stress-induced apoptosis. Aim of the present study was the identification and characterization of novel sulforaphane targets in cardiomyocytes applying a proteomic approach. Two-dimensional gel electrophoresis and mass spectrometry were used to generate protein profiles of primary neonatal rat cardiomyocytes treated and untreated with 5 µM sulforaphane for 1-48 h. According to image analysis, 64 protein spots were found as differentially expressed and their functional correlations were investigated using the MetaCore program. We mainly focused on 3 proteins: macrophage migration inhibitory factor (MIF), CLP36 or Elfin, and glyoxalase 1, due to their possible involvement in cardioprotection. Validation of the time-dependent differential expression of these proteins was performed by western blotting. In particular, to gain insight into the cardioprotective role of the modulation of glyoxalase 1 by sulforaphane, further experiments were performed using methylglyoxal to mimic glycative stress. Sulforaphane was able to counteract methylglyoxal-induced apoptosis, ROS production, and glycative stress, likely through glyoxalase 1 up-regulation. In this study, we reported for the first time new molecular targets of sulforaphane, such as MIF, CLP36 and glyoxalase 1. In particular, we gave new insights into the anti-glycative role of sulforaphane in cardiomyocytes, confirming its pleiotropic behavior in counteracting cardiovascular diseases.
Collapse
|
30
|
Sequeira V, Nijenkamp LLAM, Regan JA, van der Velden J. The physiological role of cardiac cytoskeleton and its alterations in heart failure. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:700-22. [PMID: 23860255 DOI: 10.1016/j.bbamem.2013.07.011] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/01/2013] [Accepted: 07/08/2013] [Indexed: 12/11/2022]
Abstract
Cardiac muscle cells are equipped with specialized biochemical machineries for the rapid generation of force and movement central to the work generated by the heart. During each heart beat cardiac muscle cells perceive and experience changes in length and load, which reflect one of the fundamental principles of physiology known as the Frank-Starling law of the heart. Cardiac muscle cells are unique mechanical stretch sensors that allow the heart to increase cardiac output, and adjust it to new physiological and pathological situations. In the present review we discuss the mechano-sensory role of the cytoskeletal proteins with respect to their tight interaction with the sarcolemma and extracellular matrix. The role of contractile thick and thin filament proteins, the elastic protein titin, and their anchorage at the Z-disc and M-band, with associated proteins are reviewed in physiologic and pathologic conditions leading to heart failure. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé
Collapse
Affiliation(s)
- Vasco Sequeira
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Louise L A M Nijenkamp
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Jessica A Regan
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands; Department of Physiology, Molecular Cardiovascular Research Program, Sarver Heart Center, University of Arizona, Tucson, AZ 85724, USA
| | - Jolanda van der Velden
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands; ICIN-Netherlands Heart Institute, The Netherlands.
| |
Collapse
|
31
|
Ito J, Takita M, Takimoto K, Maturana AD. Enigma homolog 1 promotes myogenic gene expression and differentiation of C2C12 cells. Biochem Biophys Res Commun 2013; 435:483-7. [PMID: 23680663 DOI: 10.1016/j.bbrc.2013.05.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 05/03/2013] [Indexed: 11/17/2022]
Abstract
The Enigma homolog (ENH) gene generates several splicing variants. The initially identified ENH1 possesses one PDZ and three LIM domains, whereas ENH2~4 lack the latter domains. The splicing switch from ENH1 to LIM-less ENHs occurs during development/maturation of skeletal and heart muscles. We examined for the roles of ENH splicing variants in muscle differentiation using C2C12 cells. Cells stably expressing ENH1 exhibited significantly higher MyoD and myogenin mRNA levels before differentiation and after 5 days in low serum-differentiating medium than mock-transfected cells. ENH1-stable transformants also retained the ability to exhibit elongated morphology with well-extended actin fibers following differentiation. In contrast, cells stably expressing ENH3 or ENH4 did not show myotube-like morphology or reorganization of actin fibers following culture in the differentiating medium. Transient overexpression of ENH1 using adenovirus supported the increased expression of muscle marker mRNAs and the formation of well-organized stress fibers, whereas ENH4 overexpression prevented these morphological changes. Furthermore, specific suppression of ENH1 expression by RNAi caused a significant reduction in MyoD mRNA level and blocked the morphological changes. These results suggest that ENH1 with multiple protein-protein interaction modules is essential for differentiation of striated muscles, whereas ectopic expression of LIM-less ENH disrupts normal muscle differentiation.
Collapse
Affiliation(s)
- Jumpei Ito
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | | | | | | |
Collapse
|
32
|
Horiuchi Y, Ishikawa M, Kaito N, Iijima Y, Tanabe Y, Ishiguro H, Arinami T. Experimental evidence for the involvement of PDLIM5 in mood disorders in hetero knockout mice. PLoS One 2013; 8:e59320. [PMID: 23593136 PMCID: PMC3620230 DOI: 10.1371/journal.pone.0059320] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 02/13/2013] [Indexed: 11/18/2022] Open
Abstract
Background Reports indicate that PDLIM5 is involved in mood disorders. The PDLIM5 (PDZ and LIM domain 5) gene has been genetically associated with mood disorders; it’s expression is upregulated in the postmortem brains of patients with bipolar disorder and downregulated in the peripheral lymphocytes of patients with major depression. Acute and chronic methamphetamine (METH) administration may model mania and the evolution of mania into psychotic mania or schizophrenia-like behavioral changes, respectively. Methods To address whether the downregulation of PDLIM5 protects against manic symptoms and cause susceptibility to depressive symptoms, we evaluated the effects of reduced Pdlim5 levels on acute and chronic METH-induced locomotor hyperactivity, prepulse inhibition, and forced swimming by using Pdlim5 hetero knockout (KO) mice. Results The homozygous KO of Pdlim5 is embryonic lethal. The effects of METH administration on locomotor hyperactivity and the impairment of prepulse inhibition were lower in Pdlim5 hetero KO mice than in wild-type mice. The transient inhibition of PDLIM5 (achieved by blocking the translocation of protein kinase C epsilon before the METH challenge) had a similar effect on behavior. Pdlim5 hetero KO mice showed increased immobility time in the forced swimming test, which was diminished after the chronic administration of imipramine. Chronic METH treatment increased, whereas chronic haloperidol treatment decreased, Pdlim5 mRNA levels in the prefrontal cortex. Imipramine increased Pdlim5 mRNA levels in the hippocampus. Conclusion These findings are partially compatible with reported observations in humans, indicating that PDLIM5 is involved in psychiatric disorders, including mood disorders.
Collapse
Affiliation(s)
- Yasue Horiuchi
- Department of Medical Genetics, Majors of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Maya Ishikawa
- Department of Medical Genetics, Majors of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Nobuko Kaito
- Department of Medical Genetics, Majors of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshimi Iijima
- Department of Medical Genetics, Majors of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshiko Tanabe
- Department of Medical Genetics, Majors of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiroki Ishiguro
- Department of Medical Genetics, Majors of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tadao Arinami
- Department of Medical Genetics, Majors of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- * E-mail:
| |
Collapse
|
33
|
Alp/Enigma family proteins cooperate in Z-disc formation and myofibril assembly. PLoS Genet 2013; 9:e1003342. [PMID: 23505387 PMCID: PMC3591300 DOI: 10.1371/journal.pgen.1003342] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 01/10/2013] [Indexed: 11/19/2022] Open
Abstract
The Drosophila Alp/Enigma family protein Zasp52 localizes to myotendinous junctions and Z-discs. It is required for terminal muscle differentiation and muscle attachment. Its vertebrate ortholog ZASP/Cypher also localizes to Z-discs, interacts with α-actinin through its PDZ domain, and is involved in Z-disc maintenance. Human mutations in ZASP cause myopathies and cardiomyopathies. Here we show that Drosophila Zasp52 is one of the earliest markers of Z-disc assembly, and we use a Zasp52-GFP fusion to document myofibril assembly by live imaging. We demonstrate that Zasp52 is required for adult Z-disc stability and pupal myofibril assembly. In addition, we show that two closely related proteins, Zasp66 and the newly identified Zasp67, are also required for adult Z-disc stability and are participating with Zasp52 in Z-disc assembly resulting in more severe, synergistic myofibril defects in double mutants. Zasp52 and Zasp66 directly bind to α-actinin, and they can also form a ternary complex. Our results indicate that Alp/Enigma family members cooperate in Z-disc assembly and myofibril formation; and we propose, based on sequence analysis, a novel class of PDZ domain likely involved in α-actinin binding.
Collapse
|
34
|
Biophysical Forces Modulate the Costamere and Z-Disc for Sarcomere Remodeling in Heart Failure. BIOPHYSICS OF THE FAILING HEART 2013. [DOI: 10.1007/978-1-4614-7678-8_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
35
|
Steinmetz PRH, Kraus JEM, Larroux C, Hammel JU, Amon-Hassenzahl A, Houliston E, Wörheide G, Nickel M, Degnan BM, Technau U. Independent evolution of striated muscles in cnidarians and bilaterians. Nature 2012; 487:231-4. [PMID: 22763458 PMCID: PMC3398149 DOI: 10.1038/nature11180] [Citation(s) in RCA: 165] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 05/03/2012] [Indexed: 12/22/2022]
Abstract
Striated muscles are present in bilaterian animals (for example, vertebrates, insects and annelids) and some non-bilaterian eumetazoans (that is, cnidarians and ctenophores). The considerable ultrastructural similarity of striated muscles between these animal groups is thought to reflect a common evolutionary origin. Here we show that a muscle protein core set, including a type II myosin heavy chain (MyHC) motor protein characteristic of striated muscles in vertebrates, was already present in unicellular organisms before the origin of multicellular animals. Furthermore, 'striated muscle' and 'non-muscle' myhc orthologues are expressed differentially in two sponges, compatible with a functional diversification before the origin of true muscles and the subsequent use of striated muscle MyHC in fast-contracting smooth and striated muscle. Cnidarians and ctenophores possess striated muscle myhc orthologues but lack crucial components of bilaterian striated muscles, such as genes that code for titin and the troponin complex, suggesting the convergent evolution of striated muscles. Consistently, jellyfish orthologues of a shared set of bilaterian Z-disc proteins are not associated with striated muscles, but are instead expressed elsewhere or ubiquitously. The independent evolution of eumetazoan striated muscles through the addition of new proteins to a pre-existing, ancestral contractile apparatus may serve as a model for the evolution of complex animal cell types.
Collapse
Affiliation(s)
- Patrick R H Steinmetz
- Department for Molecular Evolution and Development, Centre for Organismal Systems Biology, University of Vienna, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Miyazaki K, Ohno K, Tamura N, Sasaki T, Sato K. CLP36 and RIL recruit α-actinin-1 to stress fibers and differentially regulate stress fiber dynamics in F2408 fibroblasts. Exp Cell Res 2012; 318:1716-25. [PMID: 22659164 DOI: 10.1016/j.yexcr.2012.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 05/01/2012] [Accepted: 05/06/2012] [Indexed: 01/09/2023]
Abstract
CLP36 is a member of the ALP/Enigma protein family and has been shown to be localized to stress fibers in various cells. We previously reported that depletion of CLP36 caused loss of stress fibers in BeWo choriocarcinoma cells, but it remains unclear how CLP36 contributes to stress fiber formation. In this study, we generated CLP36-depleted F2408 fibroblasts and found that stress fibers showed abnormal non-oriented organization in these cells. In addition to CLP36, F2408 cells contained RIL, another ALP/Enigma protein, and we demonstrated that RIL could compensate for the role of CLP36 in stress fiber formation. CLP36 and RIL form a complex with α-actinin-1 and palladin. We found a strong correlation between loss of CLP36/RIL and failure of α-actinin-1 or palladin to localize on stress fibers. In addition, time lapse observation revealed that incorporation of RIL stabilizes stress fibers and that CLP36 influences the dynamic architecture of these fibers. Our findings indicate that CLP36 and RIL have a redundant role in the formation of stress fibers, but have different effects on stress fiber dynamics in F2408 cells.
Collapse
Affiliation(s)
- Kazufumi Miyazaki
- Department of Anatomy, Hamamatsu University School of Medicine, 431-3192 Shizuoka, Japan.
| | | | | | | | | |
Collapse
|
37
|
Ito J, Hashimoto T, Nakamura S, Aita Y, Yamazaki T, Schlegel W, Takimoto K, Maturana AD. Splicing transitions of the anchoring protein ENH during striated muscle development. Biochem Biophys Res Commun 2012; 421:232-8. [PMID: 22497889 DOI: 10.1016/j.bbrc.2012.03.142] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 03/28/2012] [Indexed: 12/31/2022]
Abstract
The ENH (PDLIM5) protein acts as a scaffold to tether various functional proteins at subcellular sites via PDZ and three LIM domains. Splicing of the ENH primary transcript generates various products with different repertories of protein interaction modules. Three LIM-containing ENH predominates in neonatal cardiac tissue, whereas LIM-less ENHs are abundant in adult hearts, as well as skeletal muscles. Here we examine the timing of splicing transitions of ENH gene products during postnatal heart development and C2C12 myoblast differentiation. Real-time PCR analysis shows that LIM-containing ENH1 mRNA is gradually decreased during postnatal heart development, whereas transcripts with the short exon 5 appear in the late postnatal period and continues to increase until at least one month after birth. The splicing transition from LIM-containing ENH1 to LIM-less ENHs is also observed during the early period of C2C12 differentiation. This transition correlates with the emergence of ENH transcripts with the short exon 5, as well as the expression of myogenin mRNA. In contrast, the shift from the short exon 5 to the exon 7 occurs in the late differentiation period. The timing of this late event corresponds to the appearance of mRNA for the skeletal myosin heavy chain MYH4. Thus, coordinated and stepwise splicing transitions result in the production of specific ENH transcripts in mature striated muscles.
Collapse
Affiliation(s)
- Jumpei Ito
- Department of Bioengineering, Nagaoka University of Technology, 1603-1 Kamitomioka, Nagaoka, Niigata 940-2188, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Guryanova OA, Drazba JA, Frolova EI, Chumakov PM. Actin cytoskeleton remodeling by the alternatively spliced isoform of PDLIM4/RIL protein. J Biol Chem 2011; 286:26849-59. [PMID: 21636573 DOI: 10.1074/jbc.m111.241554] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
RIL (product of PDLIM4 gene) is an actin-associated protein that has previously been shown to stimulate actin bundling by interacting with actin-cross-linking protein α-actinin-1 and increasing its affinity to filamentous actin. Here, we report that the alternatively spliced isoform of RIL, denoted here as RILaltCterm, functions as a dominant-negative modulator of RIL-mediated actin reorganization. RILaltCterm is regulated at the level of protein stability, and this protein isoform accumulates particularly in response to oxidative stress. We show that the alternative C-terminal segment of RILaltCterm has a disordered structure that directs the protein to rapid degradation in the core 20 S proteasomes. Such degradation is ubiquitin-independent and can be blocked by binding to NAD(P)H quinone oxidoreductase NQO1, a detoxifying enzyme induced by prolonged exposure to oxidative stress. We show that either overexpression of RILaltCterm or its stabilization by stresses counteracts the effects produced by full-length RIL on organization of actin cytoskeleton and cell motility. Taken together, the data suggest a mechanism for fine-tuning actin cytoskeleton rearrangement in response to stresses.
Collapse
Affiliation(s)
- Olga A Guryanova
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | |
Collapse
|
39
|
Abstract
Costameres are sub-membranous, Z-line associated structures found in striated muscle. They have been shown to have important roles in transmission of force from the sarcomere to the sarcolemma and extracellular matrix, maintaining mechanical integrity of the sarcolemma, and orchestrating mechanically related signaling. The costamere is akin to the more well-known focal adhesion complex present in most cells. The Z-line is a critical structural anchor for the sarcomere, but it is also a hot-spot for muscle cell signaling. Therefore functionally, the costamere represents a two-way signaling highway tethered between the Z-line and the extracellular matrix, relaying mechanical stress signals from outside the cell to intracellular signaling networks. In this role it can modulate myofibril growth and contraction. The major force generated by sarcomeres is transduced in the lateral direction from the sarcomere to the extracellular matrix through the costamere. Two major protein complexes have been described at the costamere: the dystrophin-glycoprotein complex and the integrin-vinculin-talin complex. The importance of these two protein complexes in striated muscle function has between demonstrated both in human disease and mouse models. Members of the dystrophin glycoprotein complex and integrins have both been reported to interact directly with filamin-C, thus linking costameric complexes with those present at the Z-line. Moreover, studies from our labs and others have shown that the Z-line proteins belonging to the PDZ-LIM domain protein family, enigma homolog (ENH) and cypher, may directly or indirectly be involved in this linkage. The following review will focus on the protein components of this linkage, their function in force transmission, and how the dysfunction or loss of proteins within these complexes contributes to muscular disease.
Collapse
|
40
|
Human balanced translocation and mouse gene inactivation implicate Basonuclin 2 in distal urethral development. Eur J Hum Genet 2011; 19:540-6. [PMID: 21368915 DOI: 10.1038/ejhg.2010.245] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
We studied a man with distal hypospadias, partial anomalous pulmonary venous return, mild limb-length inequality and a balanced translocation involving chromosomes 9 and 13. To gain insight into the etiology of his birth defects, we mapped the translocation breakpoints by high-resolution comparative genomic hybridization (CGH), using chromosome 9- and 13-specific tiling arrays to analyze genetic material from a spontaneously aborted fetus with unbalanced segregation of the translocation. The chromosome 13 breakpoint was ∼400 kb away from the nearest gene, but the chromosome 9 breakpoint fell within an intron of Basonuclin 2 (BNC2), a gene that encodes an evolutionarily conserved nuclear zinc-finger protein. The BNC2/Bnc2 gene is abundantly expressed in developing mouse and human periurethral tissues. In all, 6 of 48 unrelated subjects with distal hypospadias had nine novel nonsynonymous substitutions in BNC2, five of which were computationally predicted to be deleterious. In comparison, two of 23 controls with normal penile urethra morphology, each had a novel nonsynonymous substitution in BNC2, one of which was predicted to be deleterious. Bnc2(-/-) mice of both sexes displayed a high frequency of distal urethral defects; heterozygotes showed similar defects with reduced penetrance. The association of BNC2 disruption with distal urethral defects and the gene's expression pattern indicate that it functions in urethral development.
Collapse
|
41
|
LIM domains regulate protein kinase C activity: a novel molecular function. Cell Signal 2011; 23:928-34. [PMID: 21266195 DOI: 10.1016/j.cellsig.2011.01.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Accepted: 01/19/2011] [Indexed: 11/23/2022]
Abstract
Enigma homolog protein 1 (ENH1) acts as a scaffold that selectively associates protein kinases and transcription factors with cytoskeletal elements. ENH1 comprises an N-terminal PDZ domain and three C-terminal LIM domains. Through the LIM domains ENH1 interacts with the N-terminal region of protein kinase C βI (PKCβI). Here, we show that when ENH1 is co-expressed, PKCβI is translocated from the cytoplasm to the plasma membrane in the absence of any other stimulation. Moreover expression of ENH1 markedly increases PKCβI activity in the absence of PKC activators. A similar activation of PKCβI was observed with co-expression of Cypher1 or Enigma, but not other LIM proteins. The region including the three LIM domains of ENH1 (residues 415-591) appears to be sufficient for this PKCβI activation. Finally, interaction with ENH1 also increases the activity of PKCα and PKCγ, whereas it reduces PKCζ activity. These findings provide strong evidence that ENH1 activates conventional PKCs by directly binding through its LIM domains. Thus, LIM domains have a novel molecular function: the regulation of PKC activities in a PKC isoform-specific manner.
Collapse
|
42
|
Kennedy C, Lenahan M, Ryan M, Fanning S, Sheridan J, McNamara E, Carroll A, Sweeney T. Shiga toxin-producing Escherichia coli isolated from human and pig origin induce different gene expression profiles in human Caco-2 epithelial cells. Livest Sci 2010. [DOI: 10.1016/j.livsci.2010.06.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
43
|
Cheng H, Kimura K, Peter AK, Cui L, Ouyang K, Shen T, Liu Y, Gu Y, Dalton ND, Evans SM, Knowlton KU, Peterson KL, Chen J. Loss of enigma homolog protein results in dilated cardiomyopathy. Circ Res 2010; 107:348-56. [PMID: 20538684 DOI: 10.1161/circresaha.110.218735] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE The Z-line, alternatively termed the Z-band or Z-disc, is a highly ordered structure at the border between 2 sarcomeres. Enigma subfamily proteins (Enigma, Enigma homolog protein, and Cypher) of the PDZ-LIM domain protein family are Z-line proteins. Among the Enigma subfamily, Cypher has been demonstrated to play a pivotal role in the structure and function of striated muscle, whereas the role of Enigma homolog protein (ENH) in muscle remains largely unknown. OBJECTIVE We studied the role of Enigma homolog protein in the heart using global and cardiac-specific ENH knockout mouse models. METHODS AND RESULTS We identified new exons and splice isoforms for ENH in the mouse heart. Impaired cardiac contraction and dilated cardiomyopathy were observed in ENH null mice. Mice with cardiac specific ENH deletion developed a similar dilated cardiomyopathy. Like Cypher, ENH interacted with Calsarcin-1, another Z-line protein. Moreover, biochemical studies showed that ENH, Cypher short isoform and Calsarcin-1 are within the same protein complex at the Z-line. Cypher short isoform and Calsarcin-1 proteins are specifically downregulated in ENH null hearts. CONCLUSIONS We have identified an ENH-CypherS-Calsarcin protein complex at the Z-line. Ablation of ENH leads to destabilization of this protein complex and dilated cardiomyopathy.
Collapse
Affiliation(s)
- Hongqiang Cheng
- Department of Medicine, University of California San Diego, La Jolla, 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Krcmery J, Camarata T, Kulisz A, Simon HG. Nucleocytoplasmic functions of the PDZ-LIM protein family: new insights into organ development. Bioessays 2010; 32:100-8. [PMID: 20091751 DOI: 10.1002/bies.200900148] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recent work on the PDZ-LIM protein family has revealed that it has important activities at the cellular level, mediating signals between the nucleus and the cytoskeleton, with significant impact on organ development. We review and integrate current knowledge about the PDZ-LIM protein family and propose a new functional role, sequestering nuclear factors in the cytoplasm. Characterized by their PDZ and LIM domains, the PDZ-LIM family is comprised of evolutionarily conserved proteins found throughout the animal kingdom, from worms to humans. Combining two functional domains in one protein, PDZ-LIM proteins have wide-ranging and multi-compartmental cell functions during development and homeostasis. In contrast, misregulation can lead to cancer formation and progression. New emerging roles include interactions with integrins, T-box transcription factors, and receptor tyrosine kinases. Facilitating the assembly of protein complexes, PDZ-LIM proteins can act as signal modulators, influence actin dynamics, regulate cell architecture, and control gene transcription.
Collapse
Affiliation(s)
- Jennifer Krcmery
- Department of Pediatrics, Northwestern University, The Feinberg School of Medicine, Children's Memorial Research Center, Chicago, IL 60614, USA
| | | | | | | |
Collapse
|
45
|
CLP36 interacts with palladin in dorsal root ganglion neurons. Neurosci Lett 2010; 476:53-7. [PMID: 20381583 DOI: 10.1016/j.neulet.2010.03.081] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2010] [Revised: 03/31/2010] [Accepted: 03/31/2010] [Indexed: 11/20/2022]
Abstract
CLP36, a member of the alpha-actinin-associated LIM protein (ALP)/enigma protein family, plays a role in neurite outgrowth in the peripheral nervous system. However, the underlying molecular mechanisms are not known. In this study, we performed yeast two-hybrid screening of an E18 mouse whole-body cDNA library with CLP36 as the bait and isolated palladin as a CLP36-binding protein. Palladin is an actin-binding protein and it was shown to have a role in the extension of cortical neurons. A coimmunoprecipitation study showed that CLP36 and palladin formed a complex in the dorsal root ganglion (DRG). In addition, CLP36 and palladin were colocalized in the neurites and cell bodies of primary DRG neurons. Furthermore, sciatic nerve transection increased the expression of both CLP36 and palladin mRNAs in DRG neurons, with the increase in CLP36 mRNA being more prominent. This implies that CLP36 has a more specific role in nerve regeneration than palladin. Our results suggest that CLP36 may interact with palladin to influence neurite outgrowth during sciatic nerve regeneration.
Collapse
|
46
|
|
47
|
Yamazaki T, Wälchli S, Fujita T, Ryser S, Hoshijima M, Schlegel W, Kuroda S, Maturana AD. Splice variants of enigma homolog, differentially expressed during heart development, promote or prevent hypertrophy. Cardiovasc Res 2010; 86:374-82. [PMID: 20097676 DOI: 10.1093/cvr/cvq023] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
AIMS Proteins with a PDZ (for PSD-95, DLG, ZO-1) and one to three LIM (for Lin11, Isl-1, Mec-3) domains are scaffolding sarcomeric and cytoskeletal elements that form structured muscle fibres and provide for the link to intracellular signalling by selectively associating protein kinases, ion channels, and transcription factors with the mechanical stress-strain sensors. Enigma homolog (ENH) is a PDZ-LIM protein with four splice variants: ENH1 with an N-terminal PDZ domain and three C-terminal LIM domains and ENH2, ENH3, and ENH4 without LIM domains. We addressed the functional role of ENH alternative splicing. METHODS AND RESULTS We studied the expression of the four ENH isoforms in the heart during development and in a mouse model of heart hypertrophy. All four isoforms are expressed in the heart but the pattern of expression is clearly different between embryonic, neonatal, and adult stages. ENH1 appears as the embryonic isoform, whereas ENH2, ENH3, and ENH4 are predominant in adult heart. Moreover, alternative splicing of ENH was changed following induction of heart hypertrophy, producing an ENH isoform pattern similar to that of neonatal heart. Next, we tested a possible causal role of ENH1 and ENH4 in the development of cardiac hypertrophy. When overexpressed in rat neonatal cardiomyocytes, ENH1 promoted the expression of hypertrophy markers and increased cell volume, whereas, on the contrary, ENH4 overexpression prevented these changes. CONCLUSION Antagonistic splice variants of ENH may play a central role in the adaptive changes of the link between mechanical stress-sensing and signalling occurring during embryonic development and/or heart hypertrophy.
Collapse
Affiliation(s)
- Tomoko Yamazaki
- Fondation pour Recherches Médicales, Medical Faculty, University of Geneva, Geneva 1211, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Zheng M, Cheng H, Banerjee I, Chen J. ALP/Enigma PDZ-LIM domain proteins in the heart. J Mol Cell Biol 2009; 2:96-102. [PMID: 20042479 DOI: 10.1093/jmcb/mjp038] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Actinin-associated LIM protein (ALP) and Enigma are two subfamilies of Postsynaptic density 95, discs large and zonula occludens-1 (PDZ)-Lin-11, Isl1 and Mec-3 (LIM) domain containing proteins. ALP family members have one PDZ and one LIM domain, whereas Enigma proteins contain one PDZ and three LIM domains. Four ALP and three Enigma proteins have been identified in mammals, each having multiple splice variants and unique expression patterns. Functionally, these proteins bind through their PDZ domains to alpha-actinin and bind through their LIM domains or other internal protein interaction domains to other proteins, including signaling molecules. ALP and Enigma proteins have been implicated in cardiac and skeletal muscle structure, function and disease, neuronal function, bipolar disorder, tumor growth, platelet and epithelial cell motility and bone formation. This review will focus on recent advances in the biological roles of ALP/Enigma PDZ-LIM domain proteins in cardiac muscle and provide insights into mechanisms by which mutations in these proteins are related to human cardiac disease.
Collapse
Affiliation(s)
- Ming Zheng
- Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | | | | | | |
Collapse
|
49
|
Herrick S, Evers DM, Lee JY, Udagawa N, Pak DTS. Postsynaptic PDLIM5/Enigma Homolog binds SPAR and causes dendritic spine shrinkage. Mol Cell Neurosci 2009; 43:188-200. [PMID: 19900557 DOI: 10.1016/j.mcn.2009.10.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 10/20/2009] [Accepted: 10/29/2009] [Indexed: 12/24/2022] Open
Abstract
Dendritic spine morphology is thought to play important roles in synaptic development and plasticity, and morphological derangements in spines are correlated with several neurological disorders. Here, we identified an interaction between Spine-Associated RapGAP (SPAR), a postsynaptic protein that reorganizes actin cytoskeleton and drives dendritic spine head growth, and PDLIM5/Enigma Homolog (ENH), a PDZ-LIM (postsynaptic density-95/Discs large/zona occludens 1-Lin11/Isl-1/Mec3) family member. PDLIM5 has been implicated in susceptibility to bipolar disorder, major depression, and schizophrenia, but its function in neurological disease is poorly understood. We show that PDLIM5 is present in the postsynaptic density, where it promotes decreased dendritic spine head size and longer, filopodia-like morphology. Conversely, RNA interference against PDLIM5 or loss of PDLIM5 interaction with SPAR caused increased spine head diameter. Furthermore, PKC activation promoted delivery of PDLIM5 into dendritic spines and increased its spine colocalization with SPAR. These data reveal new postsynaptic functions for PDLIM5 in shrinkage of dendritic spines that may be relevant to its association with psychiatric illness.
Collapse
Affiliation(s)
- Scott Herrick
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC 20057-1464, USA
| | | | | | | | | |
Collapse
|
50
|
Camarata T, Krcmery J, Snyder D, Park S, Topczewski J, Simon HG. Pdlim7 (LMP4) regulation of Tbx5 specifies zebrafish heart atrio-ventricular boundary and valve formation. Dev Biol 2009; 337:233-45. [PMID: 19895804 DOI: 10.1016/j.ydbio.2009.10.039] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2008] [Revised: 10/03/2009] [Accepted: 10/22/2009] [Indexed: 01/29/2023]
Abstract
Tbx5 is involved in congenital heart disease, however, the mechanisms leading to organ malformation are greatly unknown. We hypothesized a model by which the Tbx5 binding protein Pdlim7 controls nuclear/cytoplasmic shuttling and function of the transcription factor. Using the zebrafish, we present in vivo significance for an essential role of Tbx5/Pdlim7 protein interaction in the regulation of cardiac formation. Knock-down of Pdlim7 results in a non-looped heart, strikingly reminiscent of the tbx5 heartstrings mutant phenotype. However, while misregulation of Pdlim7 and Tbx5 produce similar aberrant cardiac morphology, molecular and histological analysis uncovered that the Pdlim7 and Tbx5 cardiac phenotypes are due to opposite effects on valve development. Loss of Pdlim7 function causes no valve tissue to develop while lack of Tbx5 results in increased valve tissue. These opposing defects are evident before valve formation and are the result of distinct gene misregulation during specification of the atrio-ventricular (AV) boundary. We show that Pdlim7/Tbx5 interactions affect the expression of Tbx5 target genes nppa and tbx2b at the AV boundary, and their domains of misexpression directly correlate with the identified valve defects. These studies demonstrate that controlling the correct balance of Tbx5 activity is crucial for the specification of the AV boundary and valve formation.
Collapse
Affiliation(s)
- Troy Camarata
- Department of Pediatrics, Northwestern University, The Feinberg School of Medicine, Children's Memorial Research Center, 2300 Children's Plaza M/C 204, Chicago, IL 60614, USA
| | | | | | | | | | | |
Collapse
|