1
|
Pan C, Wang W, He Y, Yang B. Identification of CSRP1 as novel biomarker for hormone-sensitive prostate cancer by the combination of clinical and functional research. Cancer Cell Int 2025; 25:65. [PMID: 39994616 PMCID: PMC11849366 DOI: 10.1186/s12935-025-03708-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025] Open
Abstract
Prostate cancer (PCa) ranks as the second most common malignancy and the fifth leading cause of cancer-related deaths among men. A critical challenge lies in accurately identifying those patients at high risk for transitioning rapidly from hormone-sensitive PCa (HSPC) to lethal castration-resistant PCa (CRPC). In our study, we employed a multiomics approach involving bioinformatics analysis on datasets GSE2443 and GSE35988, along with proteomics studies, to discover that cysteine- and glycine-rich protein 1 (CSRP1) expression significantly impacts the progression of HSPC. This hypothesis was substantiated through experiments using PC3 and LNCaP prostate cancer cells, where we conducted scratch assays and apoptosis assays, all of which confirmed CSRP1's role in suppressing tumor growth. Furthermore, we elucidated the inhibitory effect of CSRP1 on tumors by performing xenograft experiments on castrated mice models. To solidify these findings in a clinical context, we constructed a nomogram model integrating CSRP1's immunohistochemistry data and clinical parameters from an actual patient cohort with HSPC. This model revealed that low CSRP1 expression indeed promotes the advancement towards CRPC. In conclusion, the level of CSRP1 expression can serve as a valuable biomarker for clinicians to predict disease progression in their patients. It has the potential to guide personalized clinical management and decision-making strategies, thereby contributing to more effective and targeted treatment approaches for HSPC patients.
Collapse
Affiliation(s)
- Chenxi Pan
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, 116023, PR China
| | - Wei Wang
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China
| | - Yi He
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China.
| | - Bo Yang
- Department of Urology, The Second Hospital of Dalian Medical University, No.467 Zhongshan Road, Dalian, 116023, PR China.
| |
Collapse
|
2
|
de Mattos ABM, Ribeiro-Silva JC, Fonseca-Alaniz MH, Valadão IC, da Silva ES, Krieger JE, Miyakawa AA. Cysteine and glycine-rich protein 3 (Crp3) as a critical regulator of elastolysis, inflammation, and smooth muscle cell apoptosis in abdominal aortic aneurysm development. Front Physiol 2023; 14:1252470. [PMID: 38173933 PMCID: PMC10762791 DOI: 10.3389/fphys.2023.1252470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening vascular disease for which surgical or endovascular repair are the only currently available therapeutic strategies. The development of AAA involves the breakdown of elastic fibers (elastolysis), infiltration of inflammatory cells, and apoptosis of smooth muscle cells (SMCs). However, the specific regulators governing these responses remain unknown. We previously demonstrated that Cysteine and glycine-rich protein 3 (Crp3) sensitizes SMCs to apoptosis induced by stretching. Building upon this finding, we aimed to investigate the influence of Crp3 on elastolysis and apoptosis during AAA development. Using the elastase-CaCl2 rat model, we observed an increase in Crp3 expression, aortic diameter, and a reduction in wall thickness in wild type rats. In contrast, Crp3-/- rats exhibited a decreased incidence of AAA, with minimal or no changes in aortic diameter and thickness. Histopathological analysis revealed the absence of SMC apoptosis and degradation of elastic fibers in Crp3-/- rats, accompanied by reduced inflammation and diminished proteolytic capacity in Crp3-/- SMCs and bone marrow-derived macrophages. Collectively, our findings provide evidence that Crp3 plays a crucial role in AAA development by modulating elastolysis, inflammation, and SMC apoptosis. These results underscore the potential significance of Crp3 in the context of AAA progression and offer new insights into therapeutic targets for this disease.
Collapse
Affiliation(s)
- Ana Barbosa Marcondes de Mattos
- Laboratorio de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Joao Carlos Ribeiro-Silva
- Laboratorio de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Miriam Helena Fonseca-Alaniz
- Laboratorio de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Iuri Cordeiro Valadão
- Laboratorio de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Erasmo Simão da Silva
- Divisão de Cirurgia Vascular e Endovascular, Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Jose Eduardo Krieger
- Laboratorio de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ayumi Aurea Miyakawa
- Laboratorio de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Cao Z, Wang H, Chen J, Zhang Y, Mo Q, Zhang P, Wang M, Liu H, Bao X, Sun Y, Zhang W, Yao Q. Silk-based hydrogel incorporated with metal-organic framework nanozymes for enhanced osteochondral regeneration. Bioact Mater 2023; 20:221-242. [PMID: 35702612 PMCID: PMC9163388 DOI: 10.1016/j.bioactmat.2022.05.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
Osteochondral defects (OCD) cannot be efficiently repaired due to the unique physical architecture and the pathological microenvironment including enhanced oxidative stress and inflammation. Conventional strategies, such as the control of implant microstructure or the introduction of growth factors, have limited functions failing to manage these complex environments. Here we developed a multifunctional silk-based hydrogel incorporated with metal-organic framework nanozymes (CuTA@SF) to provide a suitable microenvironment for enhanced OCD regeneration. The incorporation of CuTA nanozymes endowed the SF hydrogel with a uniform microstructure and elevated hydrophilicity. In vitro cultivation of mesenchymal stem cells (MSCs) and chondrocytes showed that CuTA@SF hydrogel accelerated cell proliferation and enhanced cell viability, as well as had antioxidant and antibacterial properties. Under the inflammatory environment with the stimulation of IL-1β, CuTA@SF hydrogel still possessed the potential to promote MSC osteogenesis and deposition of cartilage-specific extracellular matrix (ECM). The proteomics analysis further confirmed that CuTA@SF hydrogel promoted cell proliferation and ECM synthesis. In the full-thickness OCD model of rabbit, CuTA@SF hydrogel displayed successfully in situ OCD regeneration, as evidenced by micro-CT, histology (HE, S/O, and toluidine blue staining) and immunohistochemistry (Col I and aggrecan immunostaining). Therefore, CuTA@SF hydrogel is a promising biomaterial targeted at the regeneration of OCD.
Collapse
Affiliation(s)
- Zhicheng Cao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Hongmei Wang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Pharmaceutical Sciences, Binzhou Medical University, 264003, Yantai, Shandong, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Yanan Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Qingyun Mo
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Po Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Mingyue Wang
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Xueyang Bao
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| |
Collapse
|
4
|
Yu S, Zhao H, Meng H, Shi S, Cao S, Bian T, Ruan C. CSRP1 Promotes Colon Adenocarcinoma Growth and Serves as an Independent Risk Biomarker for Worse Prognosis. Genet Res (Camb) 2023; 2023:8586507. [PMID: 37113556 PMCID: PMC10129416 DOI: 10.1155/2023/8586507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Background Cysteine and Glycine Rich Protein 1 (CSRP1) belongs to the cysteine-rich protein family, which contains a unique double-zinc finger motif and is important for development and cellular differentiation. Abnormal expression of CSRP1 was reported within several malignancies such as prostate cancer and acute myeloid leukemia. Here, we explored function of CSRP1 within colon adenocarcinoma (COAD) for the first time. Methods The mRNA levels of CSRP1 in COADs were obtained from TCGA datasets. CSRP1 protein expressions in COADs were tested via immunohistochemistry staining. Patients' prognosis was evaluated using both univariate analysis and multivariate analysis. Two human COAD originated cancer cell lines, Caco-2, and HT-29, were used for cellular experiments including shRNA knockdown, proliferation assay, and migration assay. In vivo model was established using nude mice xenografts to further validate the role of CSRP1 in COAD progression. Results The mRNA levels of CSRP1 are elevated in COAD specimens from patients with more advanced tumor stages and higher Carcinoembryonic Antigen (CEA) levels. In addition, higher CSRP1 mRNA level indicates worse COAD prognosis. Consistently, higher CSRP1 protein expression is correlated with worse overall survival according to both univariate and multivariate analysis, indicating that CSRP1 is a new COAD prognostic factor. Furthermore, COAD cells transfected with CSRP1-shRNAs exhibit attenuated proliferation and migration capacities. Finally, growth of xenografts originated from CSRP1-knockdown cells is inhibited comparing to the control ones. Conclusions Expression of CSRP1 is positively correlated with COAD progression, which can promote tumor growth and migration. Higher CSRP1 can is a novel independent prognostic factor of COAD.
Collapse
Affiliation(s)
- Senlong Yu
- Department of Gastrointestinal Surgery, Zhuji People's Hospital of Zhejiang Province (Zhuji Affiliated Hospital of Shaoxing University), Zhuji 311800, China
| | - Haifeng Zhao
- Department of General Surgery, Zhuji Chinese Traditional Medical Hospital, Zhuji 311800, China
| | - Hongjie Meng
- Department of Gastrointestinal Surgery, Zhuji People's Hospital of Zhejiang Province (Zhuji Affiliated Hospital of Shaoxing University), Zhuji 311800, China
| | - Shengguang Shi
- Department of Gastrointestinal Surgery, Zhuji People's Hospital of Zhejiang Province (Zhuji Affiliated Hospital of Shaoxing University), Zhuji 311800, China
| | - Shenghui Cao
- Department of General Surgery, Zhuji Chinese Traditional Medical Hospital, Zhuji 311800, China
| | - Tianhua Bian
- Department of General Surgery, Zhuji Chinese Traditional Medical Hospital, Zhuji 311800, China
| | - Canping Ruan
- Colorectal Surgery Department, Changzheng Hospital, Shanghai 200003, China
| |
Collapse
|
5
|
Hayashi K, Horoiwa S, Mori K, Miyata H, Labios RJ, Morita T, Kobayashi Y, Yamashiro C, Higashijima F, Yoshimoto T, Kimura K, Nakagawa Y. Role of CRP2-MRTF interaction in functions of myofibroblasts. Cell Struct Funct 2023; 48:83-98. [PMID: 37164693 PMCID: PMC10721955 DOI: 10.1247/csf.23004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/18/2023] [Indexed: 05/12/2023] Open
Abstract
Inflammatory response induces phenotypic modulation of fibroblasts into myofibroblasts. Although transforming growth factor-βs (TGF-βs) evoke such transition, the details of the mechanism are still unknown. Here, we report that a LIM domain protein, cysteine-and glycine-rich protein 2 (CSRP2 [CRP2]) plays a vital role in the functional expression profile in myofibroblasts and cancer-associated fibroblasts (CAFs). Knock-down of CRP2 severely inhibits the expression of smooth muscle cell (SMC) genes, cell motility, and CAF-mediated collective invasion of epidermoid carcinoma. We elucidate the following molecular bases: CRP2 directly binds to myocardin-related transcription factors (MRTF-A/B [MRTFs]) and serum response factor (SRF) and stabilizes the MRTF/SRF/CArG-box complex to activate SMC gene expression. Furthermore, a three-dimensional structural analysis of CRP2 identifies the amino acids required for the CRP2-MRTF-A interaction. Polar amino acids in the C-terminal half (serine-152, glutamate-154, serine-155, threonine-156, threonine-157, and threonine-159 in human CRP2) are responsible for direct binding to MRTF-A. On the other hand, hydrophobic amino acids outside the consensus sequence of the LIM domain (tryptophan-139, phenylalanine-144, leucine-153, and leucine-158 in human CRP2) play a role in stabilizing the unique structure of the LIM domain.Key words: CRP2, 3D structure, myocardin-related transcription factor, myofibroblast, cancer-associated fibroblasts.
Collapse
Affiliation(s)
- Ken’ichiro Hayashi
- Department of RNA Biology and Neuroscience, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| | - Shinri Horoiwa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Kotaro Mori
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Hiroshi Miyata
- Department of Surgery, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka 541-8567, Japan
| | - Reuben Jacob Labios
- Department of RNA Biology and Neuroscience, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tsuyoshi Morita
- Department of Biology, Wakayama Medical University School of Medicine, 580 Mikazura, Wakayama 641-0011, Japan
| | - Yuka Kobayashi
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| | - Chiemi Yamashiro
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| | - Fumiaki Higashijima
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| | - Takuya Yoshimoto
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| | - Kazuhiro Kimura
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| | - Yoshiaki Nakagawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo-ku, Kyoto 606-8502, Japan
| |
Collapse
|
6
|
Cysteine-Rich LIM-Only Protein 4 (CRP4) Promotes Atherogenesis in the ApoE -/- Mouse Model. Cells 2022; 11:cells11081364. [PMID: 35456043 PMCID: PMC9032522 DOI: 10.3390/cells11081364] [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] [Received: 01/19/2022] [Revised: 03/31/2022] [Accepted: 04/09/2022] [Indexed: 01/27/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) can switch from their contractile state to a synthetic phenotype resulting in high migratory and proliferative capacity and driving atherosclerotic lesion formation. The cysteine-rich LIM-only protein 4 (CRP4) reportedly modulates VSM-like transcriptional signatures, which are perturbed in VSMCs undergoing phenotypic switching. Thus, we hypothesized that CRP4 contributes to adverse VSMC behaviours and thereby to atherogenesis in vivo. The atherogenic properties of CRP4 were investigated in plaque-prone apolipoprotein E (ApoE) and CRP4 double-knockout (dKO) as well as ApoE-deficient CRP4 wildtype mice. dKO mice exhibited lower plaque numbers and lesion areas as well as a reduced content of α-smooth muscle actin positive cells in the lesion area, while lesion-associated cell proliferation was elevated in vessels lacking CRP4. Reduced plaque volumes in dKO correlated with significantly less intra-plaque oxidized low-density lipoprotein (oxLDL), presumably due to upregulation of the antioxidant factor peroxiredoxin-4 (PRDX4). This study identifies CRP4 as a novel pro-atherogenic factor that facilitates plaque oxLDL deposition and identifies the invasion of atherosclerotic lesions by VSMCs as important determinants of plaque vulnerability. Thus, targeting of VSMC CRP4 should be considered in plaque-stabilizing pharmacological strategies.
Collapse
|
7
|
Yu L, Qiu C, Chen R. A narrative review of research advances in the study of molecular markers of airway smooth muscle cells. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:375. [PMID: 35434039 PMCID: PMC9011254 DOI: 10.21037/atm-22-800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/16/2022] [Indexed: 11/06/2022]
Abstract
Background and Objective Airway smooth muscle cells (ASMCs) are an important component of the airway. Their thickening and proliferation are important in pathological situations, such as airway remodeling in asthma, but their origin remains unclear. Therefore, characterizing molecular markers of ASMCs were sought to identify the source of increased ASMCs in asthmatic airway remodeling. Methods Articles for this review were derived from a review of the literature related to surface markers and biological properties of ASMCs and smooth muscle cells (SMCs) using PubMed, Google Scholar, and Web of Science. Key Content and Findings This review discusses several SMC molecular markers, describes the different developmental stages of SMCs that express different molecular markers, and summarizes several classical SMC molecular markers. However, the establishment of a specific molecular marker detection system for ASMCs still faces great challenges. Conclusions Although there is no recognized molecular marker detection system for ASMCs, and the study of the properties and sources of increased ASMCs in asthma airway remodeling is still in a state of exploration, the future is promising. Among the SMC markers described in this review, Myosin heavy chain 11 (MYH11) is a molecular marker for mature SMCs and Transgelin (TAGLN) is an early marker for SMC differentiation, and different molecular markers or combinations of molecular markers can be selected for the identification of the properties and sources of increased ASMCs in asthma airway remodeling according to the differentiation period and research needs.
Collapse
Affiliation(s)
- Li Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Chen Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| | - Rongchang Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital (Shenzhen People's Hospital), School of Medicine, Southern University of Science and Technology, Shenzhen Institute of Respiratory Diseases, Shenzhen, China
| |
Collapse
|
8
|
Weighill D, Ben Guebila M, Glass K, Quackenbush J, Platig J. Predicting genotype-specific gene regulatory networks. Genome Res 2022; 32:524-533. [PMID: 35193937 PMCID: PMC8896459 DOI: 10.1101/gr.275107.120] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 01/11/2022] [Indexed: 11/25/2022]
Abstract
Understanding how each person's unique genotype influences their individual patterns of gene regulation has the potential to improve our understanding of human health and development, and to refine genotype-specific disease risk assessments and treatments. However, the effects of genetic variants are not typically considered when constructing gene regulatory networks, despite the fact that many disease-associated genetic variants are thought to have regulatory effects, including the disruption of transcription factor (TF) binding. We developed EGRET (Estimating the Genetic Regulatory Effect on TFs), which infers a genotype-specific gene regulatory network for each individual in a study population. EGRET begins by constructing a genotype-informed TF-gene prior network derived using TF motif predictions, expression quantitative trait locus (eQTL) data, individual genotypes, and the predicted effects of genetic variants on TF binding. It then uses a technique known as message passing to integrate this prior network with gene expression and TF protein–protein interaction data to produce a refined, genotype-specific regulatory network. We used EGRET to infer gene regulatory networks for two blood-derived cell lines and identified genotype-associated, cell line–specific regulatory differences that we subsequently validated using allele-specific expression, chromatin accessibility QTLs, and differential ChIP-seq TF binding. We also inferred EGRET networks for three cell types from each of 119 individuals and identified cell type–specific regulatory differences associated with diseases related to those cell types. EGRET is, to our knowledge, the first method that infers networks reflective of individual genetic variation in a way that provides insight into the genetic regulatory associations driving complex phenotypes.
Collapse
Affiliation(s)
- Deborah Weighill
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | | | - Kimberly Glass
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA.,Harvard Medical School, Boston, Massachusetts 02115, USA
| | - John Quackenbush
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - John Platig
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA.,Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
9
|
Längst N, Adler J, Schweigert O, Kleusberg F, Cruz Santos M, Knauer A, Sausbier M, Zeller T, Ruth P, Lukowski R. Cyclic GMP-Dependent Regulation of Vascular Tone and Blood Pressure Involves Cysteine-Rich LIM-Only Protein 4 (CRP4). Int J Mol Sci 2021; 22:9925. [PMID: 34576086 PMCID: PMC8466836 DOI: 10.3390/ijms22189925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/09/2021] [Accepted: 08/25/2021] [Indexed: 01/14/2023] Open
Abstract
The cysteine-rich LIM-only protein 4 (CRP4), a LIM-domain and zinc finger containing adapter protein, has been implicated as a downstream effector of the second messenger 3',5'-cyclic guanosine monophosphate (cGMP) pathway in multiple cell types, including vascular smooth muscle cells (VSMCs). VSMCs and nitric oxide (NO)-induced cGMP signaling through cGMP-dependent protein kinase type I (cGKI) play fundamental roles in the physiological regulation of vascular tone and arterial blood pressure (BP). However, it remains unclear whether the vasorelaxant actions attributed to the NO/cGMP axis require CRP4. This study uses mice with a targeted deletion of the CRP4 gene (CRP4 KO) to elucidate whether cGMP-elevating agents, which are well known for their vasorelaxant properties, affect vessel tone, and thus, BP through CRP4. Cinaciguat, a NO- and heme-independent activator of the NO-sensitive (soluble) guanylyl cyclase (NO-GC) and NO-releasing agents, relaxed both CRP4-proficient and -deficient aortic ring segments pre-contracted with prostaglandin F2α. However, the magnitude of relaxation was slightly, but significantly, increased in vessels lacking CRP4. Accordingly, CRP4 KO mice presented with hypotonia at baseline, as well as a greater drop in systolic BP in response to the acute administration of cinaciguat, sodium nitroprusside, and carbachol. Mechanistically, loss of CRP4 in VSMCs reduced the Ca2+-sensitivity of the contractile apparatus, possibly involving regulatory proteins, such as myosin phosphatase targeting subunit 1 (MYPT1) and the regulatory light chain of myosin (RLC). In conclusion, the present findings confirm that the adapter protein CRP4 interacts with the NO-GC/cGMP/cGKI pathway in the vasculature. CRP4 seems to be part of a negative feedback loop that eventually fine-tunes the NO-GC/cGMP axis in VSMCs to increase myofilament Ca2+ desensitization and thereby the maximal vasorelaxant effects attained by (selected) cGMP-elevating agents.
Collapse
Affiliation(s)
- Natalie Längst
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Julia Adler
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Olga Schweigert
- Cardiovascular Systems Medicine and Molecular Translation, University Center of Cardiovascular Science, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (O.S.); (T.Z.)
- DZHK, German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Felicia Kleusberg
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Melanie Cruz Santos
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Amelie Knauer
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Matthias Sausbier
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Tanja Zeller
- Cardiovascular Systems Medicine and Molecular Translation, University Center of Cardiovascular Science, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (O.S.); (T.Z.)
- DZHK, German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, 20251 Hamburg, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, 72076 Tuebingen, Germany; (N.L.); (J.A.); (F.K.); (M.C.S.); (A.K.); (M.S.)
| |
Collapse
|
10
|
Ang MJ, Kang S, Moon C. Melatonin alters neuronal architecture and increases cysteine-rich protein 1 signaling in the male mouse hippocampus. J Neurosci Res 2020; 98:2333-2348. [PMID: 32754943 DOI: 10.1002/jnr.24708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 07/13/2020] [Accepted: 07/19/2020] [Indexed: 01/10/2023]
Abstract
Neuronal plasticity describes changes in structure, function, and connections of neurons. The hippocampus, in particular, has been shown to exhibit considerable plasticity regarding both physiological and morphological functions. Melatonin, a hormone released by the pineal gland, promotes cell survival and dendrite maturation of neurons in the newborn brain and protects against neurological disorders. In this study, we investigated the effect of exogenous melatonin on neuronal architecture and its possible mechanism in the hippocampus of adult male C57BL/6 mice. Melatonin treatment significantly increased the total length and complexity of dendrites in the apical and basal cornu ammonis (CA) 1 and in the dentate gyrus in mouse hippocampi. Spine density in CA1 apical dendrites was increased, but no significant differences in other subregions were observed. In primary cultured hippocampal neurons, the length and arborization of neurites were significantly augmented by melatonin treatment. Additionally, western blot and immunohistochemical analyses in both in vivo and in vitro systems revealed significant increases in the level of cysteine-rich protein 1 (crp-1) protein, which is known to be involved in dendritic branching in mouse hippocampal neurons after melatonin treatment. Our results suggest that exogenous melatonin leads to significant alterations of neuronal micromorphometry in the adult hippocampus, possibly via crp-1 signaling.
Collapse
Affiliation(s)
- Mary Jasmin Ang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Sohi Kang
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
11
|
Campos LCG, Ribeiro-Silva JC, Menegon AS, Barauna VG, Miyakawa AA, Krieger JE. Cyclic stretch-induced Crp3 sensitizes vascular smooth muscle cells to apoptosis during vein arterialization remodeling. Clin Sci (Lond) 2018; 132:CS20171601. [PMID: 29437853 DOI: 10.1042/cs20171601] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/26/2018] [Accepted: 01/31/2018] [Indexed: 02/28/2024]
Abstract
Vein graft failure limits the long-term patency of the saphenous vein used as a conduit for coronary artery bypass graft. Early graft adaptation involves some degree of intima hyperplasia to sustain the hemodynamic stress, but the progress to occlusion in some veins remains unclear. We have demonstrated that stretch-induced up-regulation of cysteine and glycine-rich protein 3 (Crp3) in rat jugular vein and human saphenous vein in response to arterialization. Here, we developed a Crp3-KO rat to investigate the role of Crp3 in vascular remodeling. After 28 days jugular vein arterialization, the intima layer was 3-fold thicker in the Crp3-KO that showed comparable smooth muscle cells (SMC) proliferation but an absence of early apoptosis observed in the wild-type rat (WT). We then investigated the role of Crp3 in early integrin-mediated signaling apoptosis in isolated jugular SMC. Interestingly, under basal conditions, ceramide treatment failed to induce apoptosis in both WT and Crp3-KO SMC. Under stretch, Crp3 expression increased in WT SMC and ceramide induced apoptosis. Immunoblotting analysis indicated that ceramide stretch-induced apoptosis in SMC is accompanied by a decrease in the phosphorylation status of both Fak and Akt, leading to an increase in Bax expression and caspase-3 cleavage. In contrast, ceramide failed to decrease Fak and Akt phosphorylation in Crp3-KO SMC and, therefore, there was no downstream induction of Bax expression and effector caspase-3 cleavage. Taken together, we provide evidence that stretch-induced Crp3 modulates vein remodeling in response to arterialization by sensitizing SMC to apoptosis.
Collapse
Affiliation(s)
| | | | | | | | - Ayumi Aurea Miyakawa
- Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Jose Eduardo Krieger
- Department of Cardiopneumology, Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, 05403-000, Brazil
| |
Collapse
|
12
|
Feng Y, Feng J, Zheng H, Wang W, Chen F, Yu Y, Cui J. Molecular cloning, characterization, and expression analysis of the three cysteine and glycine-rich protein genes in the Chinese fire-bellied newt Cynops orientalis. Gene 2018; 647:226-234. [PMID: 29317320 DOI: 10.1016/j.gene.2018.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/11/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023]
Abstract
The cysteine- and glycine-rich protein (CRP) family members, including the cysteine- and glycine-rich protein 1 (CSRP1), cysteine- and glycine-rich protein 2 (CSRP2), and the cysteine- and glycine-rich protein 3 (CSRP3), have exhibited various cellular functions during cell development and differentiation. However, the sequences of the three CSRP genes and their functions are still poorly understood in newts. In this study, we cloned the complete open reading frame (ORF) sequences of the three CSRP genes from the Chinese fire-bellied newt, Cynops orientalis (C. orientalis). The complete ORF sequences of Co-CSRP1, Co-CSRP2, and Co-CSRP3 were 582, 582, and 576bp, respectively, and encoded 193, 193, and 191 amino acids, respectively. The deduced amino acid sequences of the three CRP members showed high similarities with that of other species, particularly, with amphibians. Co-CSRP1 was highly expressed in the kidney, limb, and stomach, however, the expression was low in the spleen, heart, intestine, liver, and tail (P<0.05). The mRNA expression of Co-CSRP2 was higher in the kidney and heart than that in other organs (P<0.05). It was observed that Co-CSRP3 was only expressed in the heart, limb, and tail. The mRNA expression of Co-CSRP1 and Co-CSRP3 was lower in the digits in comparison to other limb segments. However, there was no significant difference of Co-CSRP2 mRNA expression in the four limb segments. The Co-CSRP1 and Co-CSRP2 mRNA expressions were significantly increased, whereas the expression of Co-CSRP3 was remarkably decreased during the limb regeneration. This study will provide useful information for further elucidating the role of Co-CSRP genes during newt limb regeneration.
Collapse
Affiliation(s)
- Yalong Feng
- Lab of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Juantao Feng
- Lab of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Hanxue Zheng
- Lab of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Wenjun Wang
- Lab of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Fulin Chen
- Lab of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an 710069, PR China; Provincial Key Laboratory of Biotechnology of Shaanxi, Xi'an 710069, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China Ministry of Education, Xi'an 710069, PR China
| | - Yuan Yu
- Lab of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an 710069, PR China; Provincial Key Laboratory of Biotechnology of Shaanxi, Xi'an 710069, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China Ministry of Education, Xi'an 710069, PR China.
| | - Jihong Cui
- Lab of Tissue Engineering, College of Life Sciences, Northwest University, Xi'an 710069, PR China; Provincial Key Laboratory of Biotechnology of Shaanxi, Xi'an 710069, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China Ministry of Education, Xi'an 710069, PR China.
| |
Collapse
|
13
|
Kamar A, Fahed AC, Shibbani K, El-Hachem N, Bou-Slaiman S, Arabi M, Kurban M, Seidman JG, Seidman CE, Haidar R, Baydoun E, Nemer G, Bitar F. A Novel Role for CSRP1 in a Lebanese Family with Congenital Cardiac Defects. Front Genet 2017; 8:217. [PMID: 29326753 PMCID: PMC5741687 DOI: 10.3389/fgene.2017.00217] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/04/2017] [Indexed: 12/12/2022] Open
Abstract
Despite an obvious role for consanguinity in congenital heart disease (CHD), most studies fail to document a monogenic model of inheritance except for few cases. We hereby describe a first-degree cousins consanguineous Lebanese family with 7 conceived children: 2 died in utero of unknown causes, 3 have CHD, and 4 have polydactyly. The aim of the study is to unveil the genetic variant(s) causing these phenotypes using next generation sequencing (NGS) technology. Targeted exome sequencing identified a heterozygous duplication in CSRP1 which leads to a potential frameshift mutation at position 154 of the protein. This mutation is inherited from the father, and segregates only with the CHD phenotype. The in vitro characterization demonstrates that the mutation dramatically abrogates its transcriptional activity over cardiac promoters like NPPA. In addition, it differentially inhibits the physical association of CSRP1 with SRF, GATA4, and with the newly described partner herein TBX5. Whole exome sequencing failed to show any potential variant linked to polydactyly, but revealed a novel missense mutation in TRPS1. This mutation is inherited from the healthy mother, and segregating only with the cardiac phenotype. Both TRPS1 and CSRP1 physically interact, and the mutations in each abrogate their partnership. Our findings add fundamental knowledge into the molecular basis of CHD, and propose the di-genic model of inheritance as responsible for such malformations.
Collapse
Affiliation(s)
- Amina Kamar
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Akl C Fahed
- Department of Genetics, Harvard Medical School, Boston, MA, United States.,Department of Medicine, Massachusetts General Hospital, Boston, MA, United States.,Division of Cardiology, Howard Hughes Medical Institute, Brigham and Women's Hospital, Boston, MA, United States
| | - Kamel Shibbani
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Nehme El-Hachem
- Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Salim Bou-Slaiman
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Mariam Arabi
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| | - Mazen Kurban
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon.,Department of Dermatology, American University of Beirut, Beirut, Lebanon.,Department of Dermatology, Columbia University, New York, NY, United States
| | - Jonathan G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, United States.,Division of Cardiology, Howard Hughes Medical Institute, Brigham and Women's Hospital, Boston, MA, United States
| | - Rachid Haidar
- Department of Surgery, American University of Beirut, Beirut, Lebanon
| | - Elias Baydoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Georges Nemer
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Fadi Bitar
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
14
|
Loss of LMOD1 impairs smooth muscle cytocontractility and causes megacystis microcolon intestinal hypoperistalsis syndrome in humans and mice. Proc Natl Acad Sci U S A 2017; 114:E2739-E2747. [PMID: 28292896 DOI: 10.1073/pnas.1620507114] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Megacystis microcolon intestinal hypoperistalsis syndrome (MMIHS) is a congenital visceral myopathy characterized by severe dilation of the urinary bladder and defective intestinal motility. The genetic basis of MMIHS has been ascribed to spontaneous and autosomal dominant mutations in actin gamma 2 (ACTG2), a smooth muscle contractile gene. However, evidence suggesting a recessive origin of the disease also exists. Using combined homozygosity mapping and whole exome sequencing, a genetically isolated family was found to carry a premature termination codon in Leiomodin1 (LMOD1), a gene preferentially expressed in vascular and visceral smooth muscle cells. Parents heterozygous for the mutation exhibited no abnormalities, but a child homozygous for the premature termination codon displayed symptoms consistent with MMIHS. We used CRISPR-Cas9 (CRISPR-associated protein) genome editing of Lmod1 to generate a similar premature termination codon. Mice homozygous for the mutation showed loss of LMOD1 protein and pathology consistent with MMIHS, including late gestation expansion of the bladder, hydronephrosis, and rapid demise after parturition. Loss of LMOD1 resulted in a reduction of filamentous actin, elongated cytoskeletal dense bodies, and impaired intestinal smooth muscle contractility. These results define LMOD1 as a disease gene for MMIHS and suggest its role in establishing normal smooth muscle cytoskeletal-contractile coupling.
Collapse
|
15
|
Straubinger J, Boldt K, Kuret A, Deng L, Krattenmacher D, Bork N, Desch M, Feil R, Feil S, Nemer M, Ueffing M, Ruth P, Just S, Lukowski R. Amplified pathogenic actions of angiotensin II in cysteine-rich LIM-only protein 4-negative mouse hearts. FASEB J 2017; 31:1620-1638. [PMID: 28138039 DOI: 10.1096/fj.201601186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022]
Abstract
LIM domain proteins have been identified as essential modulators of cardiac biology and pathology; however, it is unclear which role the cysteine-rich LIM-only protein (CRP)4 plays in these processes. In studying CRP4 mutant mice, we found that their hearts developed normally, but lack of CRP4 exaggerated multiple parameters of the cardiac stress response to the neurohormone angiotensin II (Ang II). Aiming to dissect the molecular details, we found a link between CRP4 and the cardioprotective cGMP pathway, as well as a multiprotein complex comprising well-known hypertrophy-associated factors. Significant enrichment of the cysteine-rich intestinal protein (CRIP)1 in murine hearts lacking CRP4, as well as severe cardiac defects and premature death of CRIP1 and CRP4 morphant zebrafish embryos, further support the notion that depleting CRP4 is incompatible with a proper cardiac development and function. Together, amplified Ang II signaling identified CRP4 as a novel antiremodeling factor regulated, at least to some extent, by cardiac cGMP.-Straubinger, J., Boldt, K., Kuret, A., Deng, L., Krattenmacher, D., Bork, N., Desch, M., Feil, R., Feil, S., Nemer, M., Ueffing, M., Ruth, P., Just, S., Lukowski, R. Amplified pathogenic actions of angiotensin II in cysteine-rich LIM-only protein 4 negative mouse hearts.
Collapse
Affiliation(s)
- Julia Straubinger
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Karsten Boldt
- Institute for Ophthalmic Research, Molecular Biology of Retinal Degenerations and Medical Proteome Center, University of Tübingen, Tübingen, Germany
| | - Anna Kuret
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Lisa Deng
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Diana Krattenmacher
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Nadja Bork
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Matthias Desch
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Robert Feil
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany; and
| | - Susanne Feil
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany; and
| | - Mona Nemer
- Laboratory of Cardiac Development and Differentiation, Department of Biochemistry, Immunology, and Microbiology, University of Ottawa, Ottawa, Ontario, Canada
| | - Marius Ueffing
- Institute for Ophthalmic Research, Molecular Biology of Retinal Degenerations and Medical Proteome Center, University of Tübingen, Tübingen, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Steffen Just
- Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology, and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany;
| |
Collapse
|
16
|
Johnson JL. Emerging regulators of vascular smooth muscle cell function in the development and progression of atherosclerosis. Cardiovasc Res 2014; 103:452-60. [PMID: 25053639 DOI: 10.1093/cvr/cvu171] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
After a period of relative senescence in the field of vascular smooth muscle cell (VSMC) research with particular regards to atherosclerosis, the last few years has witnessed a resurgence, with extensive research re-assessing potential molecular mechanisms and pathways that modulate VSMC behaviour within the atherosclerotic-prone vessel wall and the atherosclerotic plaque itself. Attention has focussed on the pathological contribution of VSMC in plaque calcification; systemic and local mediators such as inflammatory molecules and lipoproteins; autocrine and paracrine regulators which affect cell-cell and cell to matrix contacts alongside cytoskeletal changes. In this brief focused review, recent insights that have been gained into how a myriad of recently identified factors can influence the pathological behaviour of VSMC and their subsequent contribution to atherosclerotic plaque development and progression has been discussed. An overriding theme is the mechanisms involved in the alterations of VSMC function during atherosclerosis.
Collapse
Affiliation(s)
- Jason Lee Johnson
- Laboratory of Cardiovascular Pathology, School of Clinical Sciences, University of Bristol, Research Floor Level Seven, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| |
Collapse
|
17
|
Black KM, Masuzawa A, Hagberg RC, Khabbaz KR, Trovato ME, Rettagliati VM, Bhasin MK, Dillon ST, Libermann TA, Toumpoulis IK, Levitsky S, McCully JD. Preliminary biomarkers for identification of human ascending thoracic aortic aneurysm. J Am Heart Assoc 2013; 2:e000138. [PMID: 24231657 PMCID: PMC3886733 DOI: 10.1161/jaha.113.000138] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Human ascending thoracic aortic aneurysms (ATAAs) are life threatening and constitute a leading cause of mortality in the United States. Previously, we demonstrated that collagens α2(V) and α1(XI) mRNA and protein expression levels are significantly increased in ATAAs. Methods and Results In this report, the authors extended these preliminary studies using high‐throughput proteomic analysis to identify additional biomarkers for use in whole blood real‐time RT‐PCR analysis to allow for the identification of ATAAs before dissection or rupture. Human ATAA samples were obtained from male and female patients aged 65±14 years. Both bicuspid and tricuspid aortic valve patients were included and compared with nonaneurysmal aortas (mean diameter 2.3 cm). Five biomarkers were identified as being suitable for detection and identification of ATAAs using qRT‐PCR analysis of whole blood. Analysis of 41 samples (19 small, 13 medium‐sized, and 9 large ATAAs) demonstrated the overexpression of 3 of these transcript biomarkers correctly identified 79.4% of patients with ATAA of ≥4.0 cm (P<0.001, sensitivity 0.79, CI=0.62 to 0.91; specificity 1.00, 95% CI=0.42 to 1.00). Conclusion A preliminary transcript biomarker panel for the identification of ATAAs using whole blood qRT‐PCR analysis in men and women is presented.
Collapse
Affiliation(s)
- Kendra M Black
- Division of Cardiothoracic Surgery, Beth Israel Deaconess Medical Center, Boston, MA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Rocchiccioli S, Pelosi G, Rosini S, Marconi M, Viglione F, Citti L, Ferrari M, Trivella MG, Cecchettini A. Secreted proteins from carotid endarterectomy: an untargeted approach to disclose molecular clues of plaque progression. J Transl Med 2013; 11:260. [PMID: 24131807 PMCID: PMC3853772 DOI: 10.1186/1479-5876-11-260] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/18/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Atherosclerosis is the main cause of morbidity and mortality in Western countries and carotid plaque rupture is associated to acute events and responsible of 15-20% of all ischemic strokes. Several proteomics approaches have been up to now used to elucidate the molecular mechanisms involved in plaque formation as well as to identify markers of pathology severity for early diagnosis or target of therapy. The aim of this study was to characterize the plaque secretome. The advantage of this approach is that secretome mimics the in vivo condition and implies a reduced complexity compared to the whole tissue proteomics allowing the detection of under-represented potential biomarkers. METHODS Secretomes from carotid endarterectomy specimens of 14 patients were analyzed by a liquid chromatography approach coupled with label free mass spectrometry. Differential expression of proteins released from plaques and from their downstream distal side segments were evaluated in each specimen. Results were validated by Western blot analysis and ELISA assays. Histology and immunohistochemistry were performed to characterize plaques and to localise the molecular factors highlighted by proteomics. RESULTS A total of 463 proteins were identified and 31 proteins resulted differentially secreted from plaques and corresponding downstream segments. A clear-cut distinction in the distribution of cellular- and extracellular-derived proteins, evidently related to the higher cellularity of distal side segments, was observed along the longitudinal axis of carotid endarterectomy samples. The expressions of thrombospondin-1, vitamin D binding protein, and vinculin, as examples of extracellular and intracellular proteins, were immunohistologically compared between adjacent segments and validated by antibody assays. ELISA assays of plasma samples from 34 patients and 10 healthy volunteers confirmed a significantly higher concentration of thrombospondin-1 and vitamin D binding protein in atherosclerotic subjects. CONCLUSIONS Taking advantage of the optimized workflow, a detailed protein profile related to carotid plaque secretome has been produced which may assist and improve biomarker discovery of molecular factors in blood. Distinctive signatures of proteins secreted by adjacent segments of carotid plaques were evidenced and they may help discriminating markers of plaque complication from those of plaque growth.
Collapse
Affiliation(s)
- Silvia Rocchiccioli
- National Research Council, Institute of Clinical Physiology, Via Moruzzi, Pisa, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Proteomic analysis of aorta and protective effects of grape seed procyanidin B2 in db/db mice reveal a critical role of milk fat globule epidermal growth factor-8 in diabetic arterial damage. PLoS One 2012; 7:e52541. [PMID: 23285083 PMCID: PMC3528673 DOI: 10.1371/journal.pone.0052541] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 11/15/2012] [Indexed: 02/07/2023] Open
Abstract
Background Atherosclerosis is one of the major complications of type 2 diabetic patients (T2DM), leading to morbidity and mortality. Grape seed procyanidin B2 (GSPB2) has demonstrated protective effect against atherosclerosis, which is believed to be, at least in part, a result of its antioxidative effects. The aim of this study is to identify the target protein of GSPB2 responsible for the protective effect against atherosclerosis in patients with DM. Methods and Results GSPB2 (30 mg/kg body weight/day) were administrated to db/db mice for 10 weeks. Proteomics of the aorta extracts by iTRAQ analysis was obtained from db/db mice. The results showed that expression of 557 proteins were either up- or down-regulated in the aorta of diabetic mice. Among those proteins, 139 proteins were normalized by GSPB2 to the levels comparable to those in control mice. Among the proteins regulated by GSPB2, the milk fat globule epidermal growth factor-8 (MFG-E8) was found to be increased in serum level in T2DM patients; the serum level of MFG-E8 was positively correlated with carotid-femoral pulse wave velocity (CF-PWV). Inhibition of MFG-E8 by RNA interference significantly suppressed whereas exogenous recombinant MFG-E8 administration exacerbated atherogenesis the db/db mice. To gain more insights into the mechanism of action of MFG-E8, we investigated the effects of MFG-E8 on the signal pathway involving the extracellular signal-regulated kinase (ERK) and monocyte chemoattractant protein-1 (MCP-1). Treatment with recombinant MFG-E8 led to increased whereas inhibition of MFG-E8 to decreased expression of MCP-1 and phosphorylation of ERK1/2. Conclusion Our data suggests that MFG-E8 plays an important role in atherogenesis in diabetes through both ERK and MCP-1 signaling pathways. GSPB2, a well-studied antioxidant, significantly inhibited the arterial wall changes favoring atherogenesis in db/db mice by down-regulating MFG-E8 expression in aorta and its serum level. Measuring MFG-E8 serum level could be a useful clinical surrogate prognosticating atherogenesis in DM patients.
Collapse
|
20
|
Ma L, Yu YM, Guo Y, Hart RP, Schachner M. Cysteine- and glycine-rich protein 1a is involved in spinal cord regeneration in adult zebrafish. Eur J Neurosci 2012; 35:353-65. [PMID: 22288476 DOI: 10.1111/j.1460-9568.2011.07958.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In contrast to mammals, adult zebrafish have the ability to regrow descending axons and gain locomotor recovery after spinal cord injury (SCI). In zebrafish, a decisive factor for successful spinal cord regeneration is the inherent ability of some neurons to regrow their axons via (re)expressing growth-associated genes during the regeneration period. The nucleus of the medial longitudinal fascicle (NMLF) is one of the nuclei capable of regenerative response after SCI. Using microarray analysis with laser capture microdissected NMLF, we show that cysteine- and glycine-rich protein (CRP)1a (encoded by the csrp1a gene in zebrafish), the function of which is largely unknown in the nervous system, was upregulated after SCI. In situ hybridization confirmed the upregulation of csrp1a expression in neurons during the axon growth phase after SCI, not only in the NMLF, but also in other nuclei capable of regeneration, such as the intermediate reticular formation and superior reticular formation. The upregulation of csrp1a expression in regenerating nuclei started at 3 days after SCI and continued to 21 days post-injury, the longest time point studied. In vivo knockdown of CRP1a expression using two different antisense morpholino oligonucleotides impaired axon regeneration and locomotor recovery when compared with a control morpholino, demonstrating that CRP1a upregulation is an important part of the innate regeneration capability in injured neurons of adult zebrafish. This study is the first to demonstrate the requirement of CRP1a for zebrafish spinal cord regeneration.
Collapse
Affiliation(s)
- Liping Ma
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | | | | | | | | |
Collapse
|
21
|
Campbell AL, Shih HP, Xu J, Gross MK, Kioussi C. Regulation of motility of myogenic cells in filling limb muscle anlagen by Pitx2. PLoS One 2012; 7:e35822. [PMID: 22558231 PMCID: PMC3338778 DOI: 10.1371/journal.pone.0035822] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 03/22/2012] [Indexed: 01/09/2023] Open
Abstract
Cells of the ventrolateral dermomyotome delaminate and migrate into the limb buds where they give rise to all muscles of the limbs. The migratory cells proliferate and form myoblasts, which withdraw from the cell cycle to become terminally differentiated myocytes. The myogenic lineage colonizes pre-patterned regions to form muscle anlagen as muscle fibers are assembled. The regulatory mechanisms that control the later steps of this myogenic program are not well understood. The homeodomain transcription factor Pitx2 is expressed specifically in the muscle lineage from the migration of precursors to adult muscle. Ablation of Pitx2 results in distortion, rather than loss, of limb muscle anlagen, suggesting that its function becomes critical during the colonization of, and/or fiber assembly in, the anlagen. Microarrays were used to identify changes in gene expression in flow-sorted migratory muscle precursors, labeled by Lbx1(EGFP/+), which resulted from the loss of Pitx2. Very few genes showed changes in expression. Many small-fold, yet significant, changes were observed in genes encoding cytoskeletal and adhesion proteins which play a role in cell motility. Myogenic cells from genetically-tagged mice were cultured and subjected to live cell-tracking analysis using time-lapse imaging. Myogenic cells lacking Pitx2 were smaller, more symmetrical, and had more actin bundling. They also migrated about half of the total distance and velocity. Decreased motility may prevent myogenic cells from filling pre-patterned regions of the limb bud in a timely manner. Altered shape may prevent proper assembly of higher-order fibers within anlagen. Pitx2 therefore appears to regulate muscle anlagen development by appropriately balancing expression of cytoskeletal and adhesion molecules.
Collapse
Affiliation(s)
- Adam L. Campbell
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
| | - Hung-Ping Shih
- Department of Pediatrics, Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jun Xu
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
| | - Michael K. Gross
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon, United States of America
| |
Collapse
|
22
|
Järvinen PM, Myllärniemi M, Liu H, Moore HM, Leppäranta O, Salmenkivi K, Koli K, Latonen L, Band AM, Laiho M. Cysteine-rich protein 1 is regulated by transforming growth factor-β1 and expressed in lung fibrosis. J Cell Physiol 2012; 227:2605-12. [PMID: 21882188 DOI: 10.1002/jcp.23000] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Transforming growth factor-β (TGF-β) is a diverse cytokine regulating growth, apoptosis, differentiation, adhesion, invasion, and extracellular matrix production. Dysregulation of TGF-β is associated with fibrotic disorders and epithelial-mesenchymal transition, and has been linked with idiopathic pulmonary fibrosis (IPF). Cysteine-rich protein 1 (CRP1) is a small LIM-domain containing protein involved in smooth muscle differentiation. Here, we show that TGF-β1 increases the expression of CRP1 protein and that CRP1 levels increase in a biphasic fashion. A rapid transient (15-45 min) increase in CRP1 is followed by a subsequent, sustained increase in CRP1 a few hours afterwards that lasts several days. We find that TGF-β1 regulates the expression of CRP1 through Smad and non-conventional p38 MAPK signaling pathways in a transcription-independent manner and that the induction occurs concomitant with an increase in myofibroblast differentiation. Using CRP1 silencing by shRNA, we identify CRP1 as a novel factor mediating cell contractility. Furthermore, we localize CRP1 to fibroblastic foci in IPF lungs and find that CRP1 is significantly more expressed in IPF as compared to control lung tissue. The results show that CRP1 is a novel TGF-β1 regulated protein that is expressed in fibrotic lesions and may be relevant in the IPF disease.
Collapse
Affiliation(s)
- Päivi M Järvinen
- Molecular Cancer Biology Program and Haartman Institute, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
CRP1, a protein localized in filopodia of growth cones, is involved in dendritic growth. J Neurosci 2012; 31:16781-91. [PMID: 22090504 DOI: 10.1523/jneurosci.2595-11.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The cysteine-rich protein (CRP) family is a subgroup of LIM domain proteins. CRP1, which cross-links actin filaments to make actin bundles, is the only CRP family member expressed in the CNS with little known about its function in nerve cells. Here, we report that CRP1 colocalizes with actin in the filopodia of growth cones in cultured rat hippocampal neurons. Knockdown of CRP1 expression by short hairpin RNA interference results in inhibition of filopodia formation and dendritic growth in neurons. Overexpression of CRP1 increases filopodia formation and neurite branching, which require its actin-bundling activity. Expression of CRP1 with a constitutively active form of Cdc42, a GTPase involved in filopodia formation, increases filopodia formation in COS-7 cells, suggesting cooperation between the two proteins. Moreover, we demonstrate that neuronal activity upregulates CRP1 expression in hippocampal neurons via Ca²⁺ influx after depolarization. Ca²⁺/calmodulin-dependent protein kinase IV (CaMKIV) and cAMP response element binding protein mediate the Ca²⁺-induced upregulation of CRP1 expression. Furthermore, CRP1 is required for the dendritic growth induced by Ca²⁺ influx or CaMKIV. Together, these data are the first to demonstrate a role for CRP1 in dendritic growth.
Collapse
|