1
|
Brückner A, Brandtner A, Rieck S, Matthey M, Geisen C, Fels B, Stei M, Kusche-Vihrog K, Fleischmann BK, Wenzel D. Site-specific genetic and functional signatures of aortic endothelial cells at aneurysm predilection sites in healthy and AngII ApoE -/- mice. Angiogenesis 2024; 27:719-738. [PMID: 38965173 PMCID: PMC11564227 DOI: 10.1007/s10456-024-09933-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 06/16/2024] [Indexed: 07/06/2024]
Abstract
Aortic aneurysm is characterized by a pathological dilation at specific predilection sites of the vessel and potentially results in life-threatening vascular rupture. Herein, we established a modified "Häutchen method" for the local isolation of endothelial cells (ECs) from mouse aorta to analyze their spatial heterogeneity and potential role in site-specific disease development. When we compared ECs from aneurysm predilection sites of healthy mice with adjacent control segments we found regulation of genes related to extracellular matrix remodeling, angiogenesis and inflammation, all pathways playing a critical role in aneurysm development. We also detected enhanced cortical stiffness of the endothelium at these sites. Gene expression of ECs from aneurysms of the AngII ApoE-/- model when compared to sham animals mimicked expression patterns from predilection sites of healthy animals. Thus, this work highlights a striking genetic and functional regional heterogeneity in aortic ECs of healthy mice, which defines the location of aortic aneurysm formation in disease.
Collapse
Affiliation(s)
- Alexander Brückner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Adrian Brandtner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Sarah Rieck
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Michaela Matthey
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Caroline Geisen
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Benedikt Fels
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Marta Stei
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Kristina Kusche-Vihrog
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Bernd K Fleischmann
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Daniela Wenzel
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany.
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany.
| |
Collapse
|
2
|
Johansson L, Ringmark S, Bergquist J, Skiöldebrand E, Widgren A, Jansson A. A proteomics perspective on 2 years of high-intensity training in horses: a pilot study. Sci Rep 2024; 14:23684. [PMID: 39390056 PMCID: PMC11467344 DOI: 10.1038/s41598-024-75266-8] [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: 03/15/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024] Open
Abstract
The human plasma proteome is rather well studied, but not that of other species, including horses. The aims of this study were to (1), explore differences in plasma proteomic profile of young elite harness trotters kept under standardised conditions and subjected to two different training programmes for 2 years and (2) explore changes in proteomic profile over time during the training period. From September at age 1.5 year to March at age 2 years, 16 Standardbred horses were exposed to the same training programme. In March, high-intensity training was introduced and the horses were divided into two training groups (High and Low). Blood samples were collected at rest in December as 1.5-year-olds, July as 2-year-olds, December as 2.5-year-olds and December as 3.5-year-olds. Untargeted proteomics was performed and a hypothesis-generating approach was used in statistical analysis (t-tests). At the age of 2.5 years, the level of serotransferrin was higher in the High group (P = 0.01) and at least at one sampling occasion, proteins associated with fat metabolism, oxidant/antioxidant processes, cardiovascular responses, bone formation and inflammation were lower in High group compared to Low (P < 0.05). Analyses of changes over time revealed that levels of proteins involved in energy metabolism, red cell metabolism, circulation, oxidant/antioxidant activity, bone formation, inflammation, immune modulation and cellular and vascular damage changed (P < 0.05). The results indicate that proteomics analysis of blood plasma could be a viable tool for evaluation of exercise adaptations, performance and for health monitoring, with several potential biomarkers identified in this study.
Collapse
Affiliation(s)
- L Johansson
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P. O. Box 7023, Uppsala, 750 07, Sweden
| | - S Ringmark
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P. O. Box 7023, Uppsala, 750 07, Sweden
| | - J Bergquist
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P. O. Box 7023, Uppsala, 750 07, Sweden
- Department of Chemistry-BMC, Analytical Chemistry and Neurochemistry, Uppsala University, P. O. Box 599, Uppsala, 751 24, Sweden
| | - E Skiöldebrand
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P. O. Box 7023, Uppsala, 750 07, Sweden
| | - A Widgren
- Department of Chemistry-BMC, Analytical Chemistry and Neurochemistry, Uppsala University, P. O. Box 599, Uppsala, 751 24, Sweden
| | - A Jansson
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P. O. Box 7023, Uppsala, 750 07, Sweden.
| |
Collapse
|
3
|
Li Y, Xu K, Zhang Y, Mao H, Qiu Q, Yan Z, Liu X, Du Y, Chen Z. Identification of a basement membrane-related genes signature with immune correlation in bladder urothelial carcinoma and verification in vitro. BMC Cancer 2023; 23:1021. [PMID: 37872487 PMCID: PMC10591420 DOI: 10.1186/s12885-023-11340-0] [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: 01/11/2023] [Accepted: 08/26/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Bladder urothelial carcinoma (BLCA) is the most common genitourinary cancer and the prognosis of patients is often poor. However, studies of basement membrane-related genes (BM-related genes) in BLCA are less reported. Therefore, we established a BM-related genes signature to explore their functional and prognostic value in BLCA. METHODS In this study, a BM-related genes signature was constructed by LASSO-Cox regression analysis, and then a series of bioinformatics methods was used to assess the accuracy and validity of the signature. We constructed a nomogram for clinical application and also screened for possible therapeutic drugs. To investigate the functions and pathways affected by BM-related genes in BLCA, we performed functional enrichment analyses. In addition, we analyzed the immune cell infiltration landscape and immune checkpoint-related genes in the high and low-risk groups. Finally, we confirmed the prognostic value of BM-related genes in BLCA in vitro. RESULTS Combining multiple bioinformatics approaches, we identified a seven-gene signature. The accuracy and validity of this signature in predicting BLCA patients were confirmed by the test cohort. In addition, the risk score was strongly correlated with prognosis, immune checkpoint genes, drug sensitivity, and immune cell infiltration landscape. The risk score is an independent prognostic factor for BLCA patients. Further experiments revealed that all seven signature genes were differentially expressed between BLCA cell lines and normal bladder cells. Finally, overexpression of LAMA2 inhibited the migration and invasion ability of BLCA cell lines. CONCLUSIONS In summary, the BM-related genes signature was able to predict the prognosis of BLCA patients accurately, indicating that the BM-related genes possess great clinical value in the diagnosis and treatment of BLCA. Moreover, LAMA2 could be a potential therapeutic target, which provides new insights into the application of the BM-related genes in BLCA patients.
Collapse
Affiliation(s)
- Yanze Li
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
| | - Kai Xu
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
| | - Ye Zhang
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
| | - Hu Mao
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
| | - Qiangmin Qiu
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhiwei Yan
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China.
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China.
| | - Yang Du
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China.
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China.
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China.
- Institute of Urologic Disease, Renmin Hospital, Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
4
|
Moreno-Vílchez C, Llobera-Ris C, Torrecilla-Vall-Llossera C, Notario J, Figueras-Nart I. Lichen sclerosus and its association with cancer: a retrospective cohort study. Int J Dermatol 2023; 62:e84-e85. [PMID: 35394068 DOI: 10.1111/ijd.16218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/17/2022] [Accepted: 03/22/2022] [Indexed: 01/20/2023]
Affiliation(s)
- Carlos Moreno-Vílchez
- Department of Dermatology, Hospital Universitari de Bellvitge, IDIBELL, University of Barcelona, Barcelona, Spain
| | - Claudia Llobera-Ris
- Department of Dermatology, Hospital Universitari de Bellvitge, IDIBELL, University of Barcelona, Barcelona, Spain
| | | | - Jaime Notario
- Department of Dermatology, Hospital Universitari de Bellvitge, IDIBELL, University of Barcelona, Barcelona, Spain
| | - Ignasi Figueras-Nart
- Department of Dermatology, Hospital Universitari de Bellvitge, IDIBELL, University of Barcelona, Barcelona, Spain
| |
Collapse
|
5
|
Attili VR, Attili SK. Clinical and histopathological spectrum of genital lichen sclerosus in 133 cases: Focus on the diagnosis of pre-sclerotic disease. Indian J Dermatol Venereol Leprol 2022; 88:774-780. [PMID: 35389022 DOI: 10.25259/ijdvl_640_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 10/01/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUND Early inflammatory lesions of lichen sclerosus are histopathologically difficult to diagnose until the hallmark of the disease i.e., papillary sclerosis becomes visible in histological sections. Pre-sclerotic and late or resolved phases of the disease have not been extensively studied. METHODS We retrospectively reviewed all cases diagnosed as genital lichen sclerosus over a ten-year period from 2006 to 2016, correlating the clinical findings with the histological features. RESULTS A total of 133 cases of genital lichen sclerosus (90 males and 43 females) were identified. Both genders demonstrated a similar histological spectrum. Fifty eight (44%) cases were identified as having pre-sclerotic lichen sclerosus, 64 (48%) as having progressive disease and 11 (8%) cases were classified as fully resolved with atrophy. Asymptomatic vitiligoid lesions were identified in 19 (14%) cases of which 12 were male. Low-grade squamous cell carcinoma was seen within the areas affected by long-standing lichen sclerosus, in four patients (3%, 2 male). LIMITATIONS We studied only haematoxylin and eosin stained sections. The presence of basement membrane thickening could have been better illustrated with the periodic acid-Schiff stain. CONCLUSION The pathogenesis of lichen sclerosus probably involves an immune reaction to the basement membrane at the epidermal interface and around the adnexa. The initial band of inflammation shifts gradually downwards from the epidermal interface into the dermis destroying the vascular channels and appendages, resulting in excessive deposition of altered extracellular matrix. Basilar infiltration of lymphocytes along with a grossly vacuolated or thickened basement membrane is proposed as the characteristic diagnostic feature of the pre-sclerotic stage. Greater awareness of the clinicopathological spectrum of lichen sclerosus should enable early diagnosis and treatment, thereby preventing structural damage and possible malignant transformation in chronic cases.
Collapse
Affiliation(s)
| | - S K Attili
- Visakha Institute of Skin and Allergy, Visakhapatnam, Andhra Pradesh, India
| |
Collapse
|
6
|
ECM1 modified HF-MSCs targeting HSC attenuate liver cirrhosis by inhibiting the TGF-β/Smad signaling pathway. Cell Death Dis 2022; 8:51. [PMID: 35136027 PMCID: PMC8827057 DOI: 10.1038/s41420-022-00846-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/05/2022] [Accepted: 01/20/2022] [Indexed: 12/13/2022]
Abstract
Hair follicle-derived mesenchymal stem cells (HF-MSCs) show considerable therapeutic potential for liver cirrhosis (LC). To improve the effectiveness of naïve HF-MSC treatments on LC, we used bioinformatic tools to identify an exogenous gene targeting HSCs among the differentially expressed genes (DEGs) in LC to modify HF-MSCs. Extracellular matrix protein 1 (ECM1) was identified as a DEG that was significantly downregulated in the cirrhotic liver. Then, ECM1-overexpressing HF-MSCs (ECM1-HF-MSCs) were transplanted into mice with LC to explore the effectiveness and correlated mechanism of gene-overexpressing HF-MSCs on LC. The results showed that ECM1-HF-MSCs significantly improved liver function and liver pathological injury in LC after cell therapy relative to the other treatment groups. Moreover, we found that ECM1-HF-MSCs homed to the injured liver and expressed the hepatocyte-specific surface markers ALB, CK18, and AFP. In addition, hepatic stellate cell (HSC) activation was significantly inhibited in the cell treatment groups in vivo and in vitro, especially in the ECM1-HF-MSC group. Additionally, TGF-β/Smad signal inhibition was the most significant in the ECM1-HF-MSC group in vivo and in vitro. The findings indicate that the genetic modification of HF-MSCs with bioinformatic tools may provide a broad perspective for precision treatment of LC.
Collapse
|
7
|
Abstract
HER2 gene amplification occurs in many breast cancer patients and is associated with poor clinical prognosis. Trastuzumab is a therapeutic monoclonal antibody binding to HER2 and inhibits growth of HER2-positive breast cancer cells and used as a principal treatment for HER2-positive breast cancer. Unfortunately, some HER2-positive breast cancers eventually relapse after trastuzumab treatment. To investigate the molecular mechanism of trastuzumab resistance, we generated trastuzumab-resistant cells using a mouse model and found ECM1 protein is increased in trastuzumab-resistant cells. ECM1 was shown to increase EGFR signaling via upregulated matrix metalloproteinase 9/galectin-3/mucin pathway. To further find the novel mediators of HER2-driven signaling pathways in breast cancer, we investigated the upregulated proteins in HER2-overexpressing breast cancer cells using a proteomics approach and found that KRT19 is strongly upregulated in HER2-positive breast cancer cells and it activates HER2 signaling by binding to HER2 and stabilizes the receptor on the cell membrane. Moreover, we found that treatment of KRT19 antibody resulted in reduced cell viability of trastuzumab-resistant HER2-positive breast cancer cells as well as trastuzumab-sensitive cancer cells both in vitro and in vivo.
Collapse
|
8
|
Lobov AA, Yudintceva NM, Mittenberg AG, Shabelnikov SV, Mikhailova NA, Malashicheva AB, Khotin MG. Proteomic Profiling of the Human Fetal Multipotent Mesenchymal Stromal Cells Secretome. Molecules 2020; 25:E5283. [PMID: 33198321 PMCID: PMC7716221 DOI: 10.3390/molecules25225283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 01/08/2023] Open
Abstract
Secretome of multipotent mesenchymal stromal cells (MSCs) is actively used in biomedical applications such as alveolar bone regeneration, treatment of cardiovascular disease, and neurodegenerative disorders. Nevertheless, hMSCs have low proliferative potential and production of the industrial quantity of their secretome might be challenging. Human fetal multipotent mesenchymal stromal cells (FetMSCs) isolated from early human embryo bone marrow are easy to expand and might be a potential source for pharmaceutical substances production based on their secretome. However, the secretome of FetMSCs was not previously analyzed. Here, we describe the secretome of FetMSCs using LC-MALDI shotgun proteomics. We identified 236 proteins. Functional annotation of the identified proteins revealed their involvement in angiogenesis, ossification, regulation of apoptosis, and immune response processes, which made it promising for biomedical applications. The proteins identified in the FetMSCs secretome are involved in the same biological processes as proteins from previously described adult hMSCs secretomes. Nevertheless, many of the common hMSCs secretome components (such as VEGF, FGF, Wnt and TGF-β) have not been identified in the FetMSCs secretome.
Collapse
Affiliation(s)
| | | | | | | | | | - Anna B. Malashicheva
- Institute of Cytology of the Russian Academy of Science, 194064 St. Petersburg, Russia; (A.A.L.); (N.M.Y.); (A.G.M.); (S.V.S.); (N.A.M.); (M.G.K.)
| | | |
Collapse
|
9
|
Fairfield H, Costa S, DeMambro V, Schott C, Martins JDS, Ferron M, Vary C, Reagan MR. Targeting Bone Cells During Sexual Maturation Reveals Sexually Dimorphic Regulation of Endochondral Ossification. JBMR Plus 2020; 4:e10413. [PMID: 33210065 PMCID: PMC7657395 DOI: 10.1002/jbm4.10413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/01/2020] [Accepted: 09/22/2020] [Indexed: 11/18/2022] Open
Abstract
In endochondral ossification, chondroblasts become embedded in their matrix and become chondrocytes, which are mature cells that continue to proliferate, eventually becoming hypertrophic. Hypertrophic chondrocytes produce cartilage that is then resorbed by osteoclasts prior to bone matrix replacement via osteoblasts. Although sexually dimorphic bone phenotypes have long been characterized, specific modulation of the growth plate during a critical window in sexual maturation has not been evaluated. Here we report that specific depletion of osteocalcin‐ (OCN‐) expressing cells in vivo during sexual maturation leads to dimorphic bone phenotypes in males and females. At 6 to 8 weeks of age, OCN‐Cre;iDTR (inducible diphtheria toxin receptor‐expressing) mice were treated with diphtheria toxin (DT) for 2 weeks to deplete OCN+ cells. At the end of the study, long bones were collected for μCT and histomorphometry, and serum was collected for proteomic and lipidomic analyses. Ablation of OCN+ cells in mice leads to consistent trends for weight loss after 2 weeks of treatment. Females exhibited decreased skeletal parameters in response to OCN+ cell ablation treatment, as expected. However, OCN+ cell ablation in males uniquely displayed an expansion of hypertrophic chondrocytes, a widening of the growth plate, and an abnormal “clubbing” anatomy of the distal femur. Following DT treatment, mice from both sexes also underwent metabolic cage analysis, in which both sexes exhibited decreased energy expenditure. We conclude that skewing endochondral bone formation during longitudinal growth has a profound effect on body weight and energy expenditure with sex‐specific effects on developing bone. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Heather Fairfield
- Center for Molecular Medicine, Maine Medical Center Research Institute Scarborough ME USA
| | - Samantha Costa
- Center for Molecular Medicine, Maine Medical Center Research Institute Scarborough ME USA.,University of Maine Graduate School of Biomedical Science and Engineering Orono ME USA.,Graduate School of Biomedical Sciences and School of Medicine Tufts University Boston MA USA
| | - Victoria DeMambro
- Center for Molecular Medicine, Maine Medical Center Research Institute Scarborough ME USA.,University of Maine Graduate School of Biomedical Science and Engineering Orono ME USA
| | - Celine Schott
- Molecular Physiology Research Unit Institut de Recherches Cliniques de Montreal Montreal Quebec Canada.,Department of Medicine and Molecular Biology Programs of the Faculty of Medicine Université de Montreal Montreal Quebec Canada
| | | | - Mathieu Ferron
- Molecular Physiology Research Unit Institut de Recherches Cliniques de Montreal Montreal Quebec Canada.,Department of Medicine and Molecular Biology Programs of the Faculty of Medicine Université de Montreal Montreal Quebec Canada
| | - Calvin Vary
- Center for Molecular Medicine, Maine Medical Center Research Institute Scarborough ME USA.,University of Maine Graduate School of Biomedical Science and Engineering Orono ME USA.,Graduate School of Biomedical Sciences and School of Medicine Tufts University Boston MA USA
| | - Michaela R Reagan
- Center for Molecular Medicine, Maine Medical Center Research Institute Scarborough ME USA.,University of Maine Graduate School of Biomedical Science and Engineering Orono ME USA.,Graduate School of Biomedical Sciences and School of Medicine Tufts University Boston MA USA
| |
Collapse
|
10
|
Sadeghi Z, Kenyon JD, Richardson B, Khalifa AO, Cartwright M, Conroy B, Caplan A, Cameron MJ, Hijaz A. Transcriptomic Analysis of Human Mesenchymal Stem Cell Therapy in Incontinent Rat Injured Urethra. Tissue Eng Part A 2020; 26:792-810. [PMID: 32614683 DOI: 10.1089/ten.tea.2020.0033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Periurethral human mesenchymal stem cell (hMSC) injections are associated with functional improvement in animal models of postpartum stress urinary incontinence (SUI). However, limited data exist on the role of hMSCs in modulating gene expression in tissue repair after urethral injury. To this end, we quantified temporal gene expression modulation in hMSCs, and in injured rat urethral tissue, using RNA-seq in an animal model of SUI, over a 3-day period following urethral injury, and local hMSC injection. We injected PKH fluorescent-labeled hMSC into the periurethral space of rats following a 4 h vaginal distention (VD) (three rats per time point). Control rats underwent VD injury only, and all animals were euthanized at 12, 24, 36, 72 h postinjury. Rat urethral and vaginal tissues were frozen and sectioned. Fluorescent labeled hMSCs were distinguished from adjacent, unlabeled rat urethral tissue. RNA was prepared from hMSCs and urethral tissue obtained by laser dissection of frozen tissue sections and sequenced on an Illumina HiSeq 2500. Differentially expressed genes (DEGs) over 72 h were evaluated using a two-group t-test (p < 0.05). Our transcriptional analyses identified candidate genes involved in tissue injury that were broadly sorted by injury and exposure to hMSC throughout the first 72 h of acute phase of injury. DEGs in treated urethra, compared with untreated urethra, were functionally associated with tissue repair, angiogenesis, neurogenesis, and oxidative stress suppression. DEGs included a variety of cytokines, extracellular matrix stabilization and regeneration genes, cytokine signaling modification, cell cycle regulation, muscle differentiation, and stabilization. Moreover, our results revealed DEG changes in hMSCs (PKH-labeled) harvested from injured urethra. The expressions are related to DNA damage repair, transcription activation, stem cell regulation, cell survival, apoptosis, self-renewal, cell proliferation, migration, and injury response. Impact statement Stress urinary incontinence (SUI) affects nearly half of women over 40, resulting in reduced quality of life and increased health care cost. Development of SUI is multifactorial and strongly associated with vaginal delivery. While stem cell therapy in animal models of SUI and limited preliminary clinical trials demonstrate functional improvement of SUI, the role of stem cell therapy in modulating tissue repair is unclear impeding advanced clinical trials. Our work provides a new understanding of the transcriptional mechanisms with which human mesenchymal stem cells improve acute injury repair thus guiding the development of cell-based therapies for women with nonacute established SUI.
Collapse
Affiliation(s)
- Zhina Sadeghi
- University Hospitals Cleveland Medical Center, Urology Institute, Cleveland, Ohio, USA
| | - Jonathan D Kenyon
- Biology Department, Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Brian Richardson
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Ahmad O Khalifa
- University Hospitals Cleveland Medical Center, Urology Institute, Cleveland, Ohio, USA.,Menoufia University Faculty of Medicine, Urology, Shebin El-Kom, Egypt
| | - Michael Cartwright
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Britt Conroy
- University Hospitals Cleveland Medical Center, Urology Institute, Cleveland, Ohio, USA
| | - Arnold Caplan
- Biology Department, Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mark J Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Adonis Hijaz
- University Hospitals Cleveland Medical Center, Urology Institute, Cleveland, Ohio, USA
| |
Collapse
|
11
|
Steinhaeuser SS, Morera E, Budkova Z, Schepsky A, Wang Q, Rolfsson O, Riedel A, Krueger A, Hilmarsdottir B, Maelandsmo GM, Valdimarsdottir B, Sigurdardottir AK, Agnarsson BA, Jonasson JG, Ingthorsson S, Traustadottir GA, Oskarsson T, Gudjonsson T. ECM1 secreted by HER2-overexpressing breast cancer cells promotes formation of a vascular niche accelerating cancer cell migration and invasion. J Transl Med 2020; 100:928-944. [PMID: 32203150 DOI: 10.1038/s41374-020-0415-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023] Open
Abstract
The tumor microenvironment is increasingly recognized as key player in cancer progression. Investigating heterotypic interactions between cancer cells and their microenvironment is important for understanding how specific cell types support cancer. Forming the vasculature, endothelial cells (ECs) are a prominent cell type in the microenvironment of both normal and neoplastic breast gland. Here, we sought out to analyze epithelial-endothelial cross talk in the breast using isogenic non-tumorigenic vs. tumorigenic breast epithelial cell lines and primary ECs. The cellular model used here consists of D492, a breast epithelial cell line with stem cell properties, and two isogenic D492-derived EMT cell lines, D492M and D492HER2. D492M was generated by endothelial-induced EMT and is non-tumorigenic while D492HER2 is tumorigenic, expressing the ErbB2/HER2 oncogene. To investigate cellular cross talk, we used both conditioned medium (CM) and 2D/3D co-culture systems. Secretome analysis of D492 cell lines was performed using mass spectrometry and candidate knockdown (KD), and overexpression (OE) was done using siRNA and CRISPRi/CRISPRa technology. D492HER2 directly enhances endothelial network formation and activates a molecular axis in ECs promoting D492HER2 migration and invasion, suggesting an endothelial feedback response. Secretome analysis identified extracellular matrix protein 1 (ECM1) as potential angiogenic inducer in D492HER2. Confirming its involvement, KD of ECM1 reduced the ability of D492HER2-CM to increase endothelial network formation and induce the endothelial feedback, while recombinant ECM1 (rECM1) increased both. Interestingly, NOTCH1 and NOTCH3 expression was upregulated in ECs upon treatment with D492HER2-CM or rECM1 but not by CM from D492HER2 with ECM1 KD. Blocking endothelial NOTCH signaling inhibited the increase in network formation and the ability of ECs to promote D492HER2 migration and invasion. In summary, our data demonstrate that cancer-secreted ECM1 induces a NOTCH-mediated endothelial feedback promoting cancer progression by enhancing migration and invasion. Targeting this interaction may provide a novel possibility to improve cancer treatment.
Collapse
Affiliation(s)
- Sophie Sarah Steinhaeuser
- Department of Anatomy, Stem Cell Research Unit, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Erika Morera
- Department of Anatomy, Stem Cell Research Unit, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Zuzana Budkova
- Department of Anatomy, Stem Cell Research Unit, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Alexander Schepsky
- Department of Anatomy, Stem Cell Research Unit, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Qiong Wang
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland
| | - Ottar Rolfsson
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland
| | - Angela Riedel
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120, Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Aileen Krueger
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120, Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Bylgja Hilmarsdottir
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Gunhild Mari Maelandsmo
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Bryndis Valdimarsdottir
- Department of Anatomy, Stem Cell Research Unit, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Anna Karen Sigurdardottir
- Department of Anatomy, Stem Cell Research Unit, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Bjarni Agnar Agnarsson
- Department of Pathology, Landspitali-University Hospital, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Jon Gunnlaugur Jonasson
- Department of Pathology, Landspitali-University Hospital, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Saevar Ingthorsson
- Department of Anatomy, Stem Cell Research Unit, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Gunnhildur Asta Traustadottir
- Department of Anatomy, Stem Cell Research Unit, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Thordur Oskarsson
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120, Heidelberg, Germany.,Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Thorarinn Gudjonsson
- Department of Anatomy, Stem Cell Research Unit, Biomedical Center, Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland. .,Center for Systems Biology, University of Iceland, Reykjavik, Iceland. .,Department of Laboratory Hematology, Landspitali-University Hospital, Reykjavik, Iceland.
| |
Collapse
|
12
|
Zhang J, Sun W, Kong X, Zhang Y, Yang HJ, Ren C, Jiang Y, Chen M, Chen X. Mutant p53 antagonizes p63/p73-mediated tumor suppression via Notch1. Proc Natl Acad Sci U S A 2019; 116:24259-24267. [PMID: 31712410 PMCID: PMC6883818 DOI: 10.1073/pnas.1913919116] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
p53 is the most frequently mutated gene in human cancers and mutant p53 has a gain of function (GOF) that promotes tumor progression and therapeutic resistance. One of the major GOF activities of mutant p53 is to suppress 2 other p53 family proteins, p63 and p73. However, the molecular basis is not fully understood. Here, we examined whether mutant p53 antagonizes p63/p73-mediated tumor suppression in vivo by using mutant p53-R270H knockin and TAp63/p73-deficient mouse models. We found that knockin mutant p53-R270H shortened the life span of p73+/- mice and subjected TAp63+/- or p73+/- mice to T lymphoblastic lymphomas (TLBLs). To unravel the underlying mechanism, we showed that mutant p53 formed a complex with Notch1 intracellular domain (NICD) and antagonized p63/p73-mediated repression of HES1 and ECM1. As a result, HES1 and ECM1 were overexpressed in TAp63+/- ;p53R270H/- and p73+/- ;p53R270H/- TLBLs, suggesting that normal function of HES1 and ECM1 in T cell activation is hyperactivated, leading to lymphomagenesis. Together, our data reveal a previously unappreciated mechanism by which GOF mutant p53 hijacks the p63/p73-regulated transcriptional program via the Notch1 pathway.
Collapse
Affiliation(s)
- Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616;
| | - Wenqiang Sun
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Xiangmudong Kong
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Yanhong Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Hee Jung Yang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Cong Ren
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Yuqian Jiang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616
| | - Mingyi Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, CA 95616;
| |
Collapse
|
13
|
Moodley YP, Corte TJ, Oliver BG, Glaspole IN, Livk A, Ito J, Peters K, Lipscombe R, Casey T, Tan DBA. Analysis by proteomics reveals unique circulatory proteins in idiopathic pulmonary fibrosis. Respirology 2019; 24:1111-1114. [PMID: 31393655 DOI: 10.1111/resp.13668] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 04/13/2019] [Accepted: 06/04/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND OBJECTIVE Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic disease that has a poor 3-year median survival rate with unclear pathophysiology. Radiological features include bibasal, subpleural fibrosis and honeycombing while its pathology is characterized by fibroblastic foci and honeycombing. Proteomic analysis of circulating molecules in plasma may identify factors that characterize IPF and may assist in the diagnosis, prognostication and determination of pathogenic pathways in this condition. METHODS Two independent quantitative proteomic techniques were used, isobaric tags for relative and absolute quantitation (iTRAQ) and multiple reaction monitoring (MRM), to identify differentially expressed plasma proteins in a group of IPF patients in comparison to healthy controls with normal lung function matched for age and gender. RESULTS Five proteins were identified to be differentially expressed in IPF compared to healthy controls (upregulation of platelet basic protein and downregulation of actin, cytoplasmic 2, antithrombin-III, extracellular matrix protein-1 and fibronectin). CONCLUSION This study further validates the combinational use of non-targeted discovery proteomics (iTRAQ) with targeted quantitation by mass spectrometry (MRM) of soluble biomarkers to identify potentially important molecules and pathways for pulmonary diseases such as IPF.
Collapse
Affiliation(s)
- Yuben P Moodley
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia.,Stem Cell Unit, Institute for Respiratory Health, Perth, WA, Australia.,Department of Respiratory Medicine, Fiona Stanley Hospital, Perth, WA, Australia.,National Health and Medical Research Council Centre of Research Excellence in Pulmonary Fibrosis, University of Sydney, Sydney, NSW, Australia
| | - Tamera J Corte
- National Health and Medical Research Council Centre of Research Excellence in Pulmonary Fibrosis, University of Sydney, Sydney, NSW, Australia.,Department of Respiratory Medicine, Royal Prince Alfred Hospital, Sydney, NSW, Australia.,School of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Brian G Oliver
- Woolcock Institute of Medical Research, University of Sydney, Sydney, NSW, Australia.,Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Ian N Glaspole
- National Health and Medical Research Council Centre of Research Excellence in Pulmonary Fibrosis, University of Sydney, Sydney, NSW, Australia.,Department of Allergy and Respiratory Medicine, The Alfred Hospital, Melbourne, VIC, Australia.,Faculty of Medicine, Monash University, Melbourne, VIC, Australia
| | | | - Jason Ito
- Proteomics International, Perth, WA, Australia
| | | | | | - Tammy Casey
- Proteomics International, Perth, WA, Australia
| | - Dino B A Tan
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia.,Stem Cell Unit, Institute for Respiratory Health, Perth, WA, Australia
| |
Collapse
|
14
|
Yu VZ, Ko JMY, Ning L, Dai W, Law S, Lung ML. Endoplasmic reticulum-localized ECM1b suppresses tumor growth and regulates MYC and MTORC1 through modulating MTORC2 activation in esophageal squamous cell carcinoma. Cancer Lett 2019; 461:56-64. [PMID: 31319137 DOI: 10.1016/j.canlet.2019.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/06/2019] [Accepted: 07/11/2019] [Indexed: 12/29/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a deadly disease with dismal 5-year survival. Extracellular matrix protein 1 (ECM1) was identified as one of the most downregulated genes by transcriptomic analysis of normal esophageal/ESCC paired tissue samples. ECM1 plays oncogenic roles in cancer development in various cancer types. However, little is known about its role in ESCC. In vivo and in vitro functional assays coupled with analyses on public datasets and detailed molecular and mechanistic analyses were used to study the gene. We demonstrate that as opposed to the previously identified oncogenic role of ECM1a, ECM1b is a novel tumor suppressor in ESCC. ECM1 is significantly downregulated in ESCC and several other squamous cell carcinomas. ECM1b encodes a cellular protein that suppresses MYC protein expression and MTORC1 signaling activity. MTORC2 inactivation leads to suppressed MYC expression and MTORC1 signaling. ECM1b localizes to the endoplasmic reticulum and suppresses MTORC2 activation by inhibiting MTORC2/ribosome association. By regulating MTORC2/MYC/MTORC1 signaling, ECM1b suppresses general protein translation and enhances chemosensitivity. We provide evidence establishing a novel role of ECM1 in cancer that suggests ECM1b as a biomarker for ESCC disease management.
Collapse
Affiliation(s)
- Valen Zhuoyou Yu
- Department of Clinical Oncology, University of Hong Kong Li Ka Shing Faculty of Medicine, Pokfulam, Hong Kong
| | - Josephine Mun Yee Ko
- Department of Clinical Oncology, University of Hong Kong Li Ka Shing Faculty of Medicine, Pokfulam, Hong Kong
| | - Lvwen Ning
- Department of Clinical Oncology, University of Hong Kong Li Ka Shing Faculty of Medicine, Pokfulam, Hong Kong
| | - Wei Dai
- Department of Clinical Oncology, University of Hong Kong Li Ka Shing Faculty of Medicine, Pokfulam, Hong Kong
| | - Simon Law
- Department of Surgery, University of Hong Kong Li Ka Shing Faculty of Medicine, Pokfulam, Hong Kong
| | - Maria Li Lung
- Department of Clinical Oncology, University of Hong Kong Li Ka Shing Faculty of Medicine, Pokfulam, Hong Kong.
| |
Collapse
|
15
|
Ghazawi FM, Proulx ESC, Jafarian F. A novel nonsense mutation in exon 9 in the extracellular matrix protein 1 gene associated with lipoid proteinosis: A case report. SAGE Open Med Case Rep 2019; 7:2050313X19850359. [PMID: 31205714 PMCID: PMC6537054 DOI: 10.1177/2050313x19850359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Lipoid proteinosis is a rare autosomal recessive genodermatosis that is caused by
loss-of-function mutations in the extracellular matrix protein 1 gene. This
study identifies a novel nonsense mutation in exon 9 of the extracellular matrix
protein 1 gene associated with lipoid proteinosis, contributing to recent
advances in our understanding of the molecular genetics underlying this disease.
It is important to identify the mutations in the extracellular matrix protein 1
gene that are associated with lipoid proteinosis and how these affect protein
function. Understanding the molecular basis for such genetic disorders may lead
to novel therapeutic approaches for treating hereditary genodermatoses.
Collapse
Affiliation(s)
- Feras M Ghazawi
- Division of Dermatology, University of Ottawa, Ottawa, ON, Canada
| | - Etienne Saint-Cyr Proulx
- Hôpital de la Cité-de-la-Santé, Laval, QC, Canada.,Innovaderm Research Inc., Montreal, QC, Canada
| | - Fatemeh Jafarian
- Division of Pediatric Dermatology, McGill University Health Center, Montreal Children's Hospital, Montreal, QC, Canada
| |
Collapse
|
16
|
Hardy SA, Mabotuwana NS, Murtha LA, Coulter B, Sanchez-Bezanilla S, Al-Omary MS, Senanayake T, Loering S, Starkey M, Lee RJ, Rainer PP, Hansbro PM, Boyle AJ. Novel role of extracellular matrix protein 1 (ECM1) in cardiac aging and myocardial infarction. PLoS One 2019; 14:e0212230. [PMID: 30789914 PMCID: PMC6383988 DOI: 10.1371/journal.pone.0212230] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/29/2019] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION The prevalence of heart failure increases in the aging population and following myocardial infarction (MI), yet the extracellular matrix (ECM) remodeling underpinning the development of aging- and MI-associated cardiac fibrosis remains poorly understood. A link between inflammation and fibrosis in the heart has long been appreciated, but has mechanistically remained undefined. We investigated the expression of a novel protein, extracellular matrix protein 1 (ECM1) in the aging and infarcted heart. METHODS Young adult (3-month old) and aging (18-month old) C57BL/6 mice were assessed. Young mice were subjected to left anterior descending artery-ligation to induce MI, or transverse aortic constriction (TAC) surgery to induce pressure-overload cardiomyopathy. Left ventricle (LV) tissue was collected early and late post-MI/TAC. Bone marrow cells (BMCs) were isolated from young healthy mice, and subject to flow cytometry. Human cardiac fibroblast (CFb), myocyte, and coronary artery endothelial & smooth muscle cell lines were cultured; human CFbs were treated with recombinant ECM1. Primary mouse CFbs were cultured and treated with recombinant angiotensin-II or TGF-β1. Immunoblotting, qPCR and mRNA fluorescent in-situ hybridization (mRNA-FISH) were conducted on LV tissue and cells. RESULTS ECM1 expression was upregulated in the aging LV, and in the infarct zone of the LV early post-MI. No significant differences in ECM1 expression were found late post-MI or at any time-point post-TAC. ECM1 was not expressed in any resident cardiac cells, but ECM1 was highly expressed in BMCs, with high ECM1 expression in granulocytes. Flow cytometry of bone marrow revealed ECM1 expression in large granular leucocytes. mRNA-FISH revealed that ECM1 was indeed expressed by inflammatory cells in the infarct zone at day-3 post-MI. ECM1 stimulation of CFbs induced ERK1/2 and AKT activation and collagen-I expression, suggesting a pro-fibrotic role. CONCLUSIONS ECM1 expression is increased in ageing and infarcted hearts but is not expressed by resident cardiac cells. Instead it is expressed by bone marrow-derived granulocytes. ECM1 is sufficient to induce cardiac fibroblast stimulation in vitro. Our findings suggest ECM1 is released from infiltrating inflammatory cells, which leads to cardiac fibroblast stimulation and fibrosis in aging and MI. ECM1 may be a novel intermediary between inflammation and fibrosis.
Collapse
Affiliation(s)
- Sean A. Hardy
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Nishani S. Mabotuwana
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Lucy A. Murtha
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Brianna Coulter
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Sonia Sanchez-Bezanilla
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Mohammed S. Al-Omary
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Tharindu Senanayake
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
| | - Svenja Loering
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Malcolm Starkey
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
| | - Randall J. Lee
- Department of Medicine, Division of Cardiology, University of California San Francisco, San Francisco, CA, United States of America
- Edyth and Eli Broad Center for Regenerative Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States of America
| | - Peter P. Rainer
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Philip M. Hansbro
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre’s for Healthy Lungs and GrowUpWell, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Centre for inflammation, Centenary Institute, Sydney, NSW, Australia
- University of Technology, Faculty of Science, Ultimo, NSW, Australia
| | - Andrew J. Boyle
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia
- * E-mail:
| |
Collapse
|
17
|
Wang Z, Zhou Q, Li A, Huang W, Cai Z, Chen W. Extracellular matrix protein 1 (ECM1) is associated with carcinogenesis potential of human bladder cancer. Onco Targets Ther 2019; 12:1423-1432. [PMID: 30863109 PMCID: PMC6389008 DOI: 10.2147/ott.s191321] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Bladder cancer (BCa) is a common urological malignant tumor worldwide, and recurrence and death still remain high. New therapeutic targets are needed to treat patients who are not sensitive to current therapy. Extracellular matrix protein 1 (ECM1) is a key player in multiple epithelial malignancies. However, the knowledge regarding the expression of ECM1 in BCa and the mechanisms by which ECM1 affects BCa tumor progression is unclear. Materials and methods ECM1 expression levels in BCa tissues and cells were detected by quantitative real-time PCR (qRT-PCR), immunohistochemistry and Western blot. ECM1 expression was suppressed by shRNAs. Cell Counting Kit-8 (CCK-8), luminescent cell viability assay and 5-ethynyl-2′-deoxyuridine (EdU) assay were used to detect cell proliferation. Flow cytometry and transwell assay were used to evaluate cell apoptosis and invasion, respectively. All statistical analyses were performed by using the GraphPad Prism 7 software package. Results In this study, the expression of ECM1 in BCa specimens and cell lines was examined and displayed a significant increase compared with noncancerous counterparts, while ECM1-knockdown affected not only cell proliferation and migration, but also cell invasion ability and apoptosis potential, corresponding to the finding that ECM1 overexpression in BCa patients was associated with a poor prognosis. Additionally, after suppression of ECM1, the expression of glucose transporter 1 (GLUT1), lactate dehydrogenase (LDHA) and hypoxia-inducible factor 1α (HIF-1α), genes involved in Warburg effect regulation, were significantly decreased, and the lactate production was also obviously reduced in ECM1-silenced cells. Conclusion Our investigations revealed that the expression of ECM1 was closely associated with tumor cell growth, migration and apoptosis at least in part through regulation of Warburg effect, defining ECM1 as an effective predictor in the carcinogenesis and postoperative recurrence of human BCa.
Collapse
Affiliation(s)
- Zhicai Wang
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ;
| | - Qun Zhou
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ;
| | - Aolin Li
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ;
| | - Weiren Huang
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ; .,Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China, .,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China,
| | - Zhiming Cai
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ; .,Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China, .,Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China,
| | - Wei Chen
- Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China, ;
| |
Collapse
|
18
|
Abstract
The cellular microenvironment often plays a crucial role in disease development and progression. In recessive dystrophic epidermolysis bullosa (RDEB), biallelic mutations of the gene COL7A1, encoding for collagen VII, the main component of anchoring fibrils, lead to a loss of collagen VII in the extracellular matrix (ECM). Loss of collagen VII in skin is linked to a destabilization of the dermal-epidermal junction zone, blister formation, chronic wounds, fibrosis, and aggressive skin cancer. Thus, RDEB cells can serve as a model system to study the effects of a perturbed ECM on the cellular proteome. In this chapter, we describe in detail the combination of stable isotope labeling by amino acids in cell culture (SILAC) of primary skin fibroblasts with reseeding of fibroblasts on decellularized collagen VII-positive and -negative ECM to study the consequences of collagen VII loss on the cellular proteome. This approach allows the quantitative, time-resolved analysis of cellular protein dynamics in response to ECM perturbation by liquid chromatography-mass spectrometry.
Collapse
|
19
|
Poma AM, Giannini R, Piaggi P, Ugolini C, Materazzi G, Miccoli P, Vitti P, Basolo F. A six-gene panel to label follicular adenoma, low- and high-risk follicular thyroid carcinoma. Endocr Connect 2018; 7:124-132. [PMID: 29298844 PMCID: PMC5754511 DOI: 10.1530/ec-17-0261] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/27/2017] [Indexed: 12/19/2022]
Abstract
The distinction between follicular thyroid carcinomas (FTCs) and follicular-patterned benign lesions is almost impossible on fine-needle aspiration cytology. Furthermore, minimally invasive FTCs (MI-FTCs) with less than 4 vascular invasion foci generally have an excellent prognosis, but there are exceptions and, so far, no molecular marker appears able to identify them reliably. We aimed to distinguish benign lesions from low- and high-risk FTCs by a small-scale combination of genes. The expression analysis of 75 selected genes was performed on 18 follicular adenomas (FAs), 14 MI-FTCs and 6 widely invasive FTC (WI-FTCs). The mutational status of the RAS genes, TERT promoter and PAX8-PPARG rearrangements was also investigated. Seven samples were mutated, namely 3 MI-FTCs and 4 WI-FTCs. Twenty-five genes were differentially expressed (FDR <0.05) between FAs and WI-FTCs. Six of these (ECM1, RXRG, SDPR, SLC26A4, TIFF3, TIMP1) were also differently expressed among MI-FTCs and FAs or WI-FTCs and were considered to build a classification model, which was tested to classify samples according to their histological class. Hence, 31 out of 38 were correctly classified, and accuracy remained high after cross-validation (27/38). The 2 MI-FTCs incorrectly classified as WI-FTCs harbored both RAS and TERT promoter mutations. The capability of these six genes to stratify benign, low- and high-risk lesions appears to be promising in supporting the diagnosis of indeterminate thyroid nodules.
Collapse
Affiliation(s)
- Anello Marcello Poma
- Department of SurgicalMedical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Riccardo Giannini
- Department of SurgicalMedical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Paolo Piaggi
- National Institute of Diabetes and Digestive and Kidney DiseasesNational Institutes of Health, Phoenix, Arizona, USA
| | - Clara Ugolini
- Department of Laboratory MedicineSection of Pathology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Gabriele Materazzi
- Department of SurgicalMedical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Paolo Miccoli
- Department of SurgicalMedical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Paolo Vitti
- Department of Clinical and Experimental MedicineUniversity of Pisa, Pisa, Italy
| | - Fulvio Basolo
- Department of SurgicalMedical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| |
Collapse
|
20
|
Gan L, Meng J, Xu M, Liu M, Qi Y, Tan C, Wang Y, Zhang P, Weng W, Sheng W, Huang M, Wang Z. Extracellular matrix protein 1 promotes cell metastasis and glucose metabolism by inducing integrin β4/FAK/SOX2/HIF-1α signaling pathway in gastric cancer. Oncogene 2017; 37:744-755. [PMID: 29059156 DOI: 10.1038/onc.2017.363] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/03/2017] [Accepted: 08/25/2017] [Indexed: 12/18/2022]
Abstract
Extracellular matrix protein 1 (ECM1) is related to strong invasiveness and poor prognosis in major malignancies, but the underlying mechanism remains unknown. Here we aimed to elucidate the function of ECM1 on cell metastasis and glucose metabolism in gastric cancer (GC). The level of ECM1 in sera and tissues of patient with GC were positively correlated with tumor invasion and recurrence. Genetic manipulation of ECM1 expression affected cell metastasis and glucose metabolism in GC cell lines. Enhanced ECM1 expression facilitated gene expression levels associated with epithelial-mesenchymal transition (EMT) and glucose metabolism. Interestingly, our results indicated that ECM1 directly interacted with integrin β4 (ITGB4) and activated ITGB4/focal adhesion kinase (FAK)/glycogen synthase kinase 3β signaling pathway, which further induced the expression of transcription factor SOX2. Aberrant expression of SOX2 altered gene expression of EMT factors and glucose metabolism enzymes. Furthermore, SOX2 enhanced hypoxia-inducible factor α (HIF-1α) promoter activity to regulate glucose metabolism. The micro-positron emission tomography/computed tomography imaging of xenograft model showed that ECM1 substantially increased 18F-fluorodeoxyglucose uptake in xenograft tumors. Using in vivo mouse tail vein injection experiments, ECM1 was also found to increase in lung surface metastasis. These findings provide evidence that ECM1 regulates GC cell metastasis and glucose metabolism by inducing ITGB4/FAK/SOX2/HIF-1α signal pathway and have important implications for the development of therapeutic target to prevent tumor metastasis and recurrence.
Collapse
Affiliation(s)
- L Gan
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - J Meng
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - M Xu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - M Liu
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Y Qi
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - C Tan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Y Wang
- Nanchang Medical College, Nanchang University, Nanchang, China
| | - P Zhang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - W Weng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - W Sheng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - M Huang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Z Wang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Philley JV, Kannan A, Griffith DE, Devine MS, Benwill JL, Wallace RJ, Brown-Elliott BA, Thakkar F, Taskar V, Fox JG, Alqaid A, Bains H, Gupta S, Dasgupta S. Exosome secretome and mediated signaling in breast cancer patients with nontuberculous mycobacterial disease. Oncotarget 2017; 8:18070-18081. [PMID: 28160560 PMCID: PMC5392308 DOI: 10.18632/oncotarget.14964] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 01/10/2017] [Indexed: 12/03/2022] Open
Abstract
Bronchiectasis Nontuberculous mycobacterium (NTMnb) infection is an emerging health problem in breast cancer (BCa) patients. We measured sera exosome proteome in BCa-NTMnb subjects and controls by Mass Spectroscopy. Extracellular matrix protein 1 (ECM1) was detected exclusively in the circulating exosomes of 82% of the BCa-NTMnb cases. Co-culture of ECM1+ exosomes with normal human mammary epithelial cells induced epithelial to mesenchymal transition accompanied by increased Vimentin/CDH1 expression ratio and Glutamate production. Co-culture of the ECM1+ exosomes with normal human T cells modulated their cytokine production. The ECM1+ exosomes were markedly higher in sera obtained from BCa-NTMnb subjects. Exclusive expression of APN, APOC4 and AZGP1 was evident in the circulating exosomes of these BCa-NTMnb cases, which predicts disease prevalence independent of the body max index in concert with ECM1. Monitoring ECM1, APN, APOC4 and AZGP1 in the circulating exosomes could be beneficial for risk assessment, monitoring and surveillance of BCa-NTMnb.
Collapse
Affiliation(s)
- Julie V Philley
- Department of Medicine, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Anbarasu Kannan
- Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Texas, USA
| | - David E Griffith
- Department of Medicine, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Megan S Devine
- Department of Medicine, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Jeana L Benwill
- Department of Medicine, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Richard J Wallace
- Department of Medicine, The University of Texas Health Science Center at Tyler, Texas, USA.,The Mycobacteria/Nocardia Research Laboratory Department of Microbiology, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Barbara A Brown-Elliott
- The Mycobacteria/Nocardia Research Laboratory Department of Microbiology, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Foram Thakkar
- Department of Medicine, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Varsha Taskar
- Department of Medicine, The University of Texas Health Science Center at Tyler, Texas, USA
| | - James G Fox
- Department of Medicine, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Ammar Alqaid
- Department of Medicine, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Hernaina Bains
- Department of Medicine, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Sudeep Gupta
- Medical Oncology, Tata Memorial Center, Mumbai, India
| | - Santanu Dasgupta
- Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Texas, USA
| |
Collapse
|
22
|
Sanchez C, Bay-Jensen AC, Pap T, Dvir-Ginzberg M, Quasnichka H, Barrett-Jolley R, Mobasheri A, Henrotin Y. Chondrocyte secretome: a source of novel insights and exploratory biomarkers of osteoarthritis. Osteoarthritis Cartilage 2017; 25:1199-1209. [PMID: 28232143 DOI: 10.1016/j.joca.2017.02.797] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/31/2017] [Accepted: 02/14/2017] [Indexed: 02/02/2023]
Abstract
The extracellular matrix (ECM) of articular cartilage is comprised of complex networks of proteins and glycoproteins, all of which are expressed by its resident cell, the chondrocyte. Cartilage is a unique tissue given its complexity and ability to resist repeated load and deformation. The mechanisms by which articular cartilage maintains its integrity throughout our lifetime is not fully understood, however there are numerous regulatory pathways known to govern ECM turnover in response to mechanical stimuli. To further our understanding of this field, we envision that proteomic analysis of the secretome will provide information on how the chondrocyte remodels the surrounding ECM in response to load, in addition to providing information on the metabolic state of the cell. In this review, we attempt to summarize the recent mass spectrometry-based proteomic discoveries in healthy and diseased cartilage and chondrocytes, to facilitate the discovery of novel biomarkers linked to degenerative pathologies, such as osteoarthritis (OA).
Collapse
Affiliation(s)
- C Sanchez
- Bone and Cartilage Research Unit, Arthropôle Liège, University of Liège, CHU Sart-Tilman, Belgium; The D-BOARD European Consortium for Biomarker Discovery.
| | - A-C Bay-Jensen
- The D-BOARD European Consortium for Biomarker Discovery; Department of Rheumatology, Biomarkers and Research, Nordic Bioscience, Herlev Hovedgade 207, 2730, Herlev, Denmark.
| | - T Pap
- The D-BOARD European Consortium for Biomarker Discovery; Institute of Experimental Musculoskeletal Medicine, University Hospital Munster, Domagkstrasse 3, D-48149, Munster, Germany.
| | - M Dvir-Ginzberg
- The D-BOARD European Consortium for Biomarker Discovery; Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University of Jerusalem, P.O. Box 12272, Jerusalem, 91120, Israel.
| | - H Quasnichka
- The D-BOARD European Consortium for Biomarker Discovery; Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, United Kingdom.
| | - R Barrett-Jolley
- The D-BOARD European Consortium for Biomarker Discovery; Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom.
| | - A Mobasheri
- The D-BOARD European Consortium for Biomarker Discovery; Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, United Kingdom; Faculty of Health and Medical Sciences, Duke of Kent Building, University of Surrey, Guildford, Surrey, GU2 7XH, United Kingdom; Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom; Center of Excellence in Genomic Medicine Research (CEGMR), King Fahd Medical Research Center (KFMRC), Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
| | - Y Henrotin
- Bone and Cartilage Research Unit, Arthropôle Liège, University of Liège, CHU Sart-Tilman, Belgium; The D-BOARD European Consortium for Biomarker Discovery.
| |
Collapse
|
23
|
An atlas of bloodstream-accessible bone marrow proteins for site-directed therapy of acute myeloid leukemia. Leukemia 2017; 32:510-519. [DOI: 10.1038/leu.2017.208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 06/01/2017] [Accepted: 06/20/2017] [Indexed: 12/15/2022]
|
24
|
Izadi F, Mahjoubi F, Farhadi M, Kalayinia S, Bidmeshkipour A, Tavakoli MM, Samanian S. Extracellular matrix protein 1 gene (ECM1) mutations in nine Iranian families with lipoid proteinosis. Indian J Med Res 2017; 143:303-7. [PMID: 27241643 PMCID: PMC4892076 DOI: 10.4103/0971-5916.182620] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background & objectives: Lipoid proteinosis (LP) is an autosomal recessive disease. Clinical characteristics of this disease are hoarse voice, scarring of the skin, brain calcifications, and eyelid papules (moniliform blepharosis). Mutations in the ECM1 gene on 1q21.2 are responsible for this disease. This study was conducted to investigate the mutation spectrum of ECM1 gene in nine Iranian families having at least one LP patient diagnosed clinically. Methods: The entire ECM1 gene was screened using PCR and direct sequencing in nine Iranian families with 12 suspected LP patients who were referred to the clinic, along with their parents and siblings. Thirty healthy individuals were included as controls. Results: In only one patient a homozygous G>A transition at nucleotide c.806 in exon 7 was detected. A G>A substitution at nucleotide 1243 in exon 8 that changes glycine (GGT) to serine (AGT) was observed in most of our patients. Furthermore, in one patient there was a change in the sequence of intron 8, the A>T transition in nucleotide 4307. In addition, in two cases (one patient and one healthy mother with affected child) there was a C (4249) deletion in intron 8. Interpretation & conclusions: Our results indicate that although mutation in ECM1gene is responsible for lipoid proteinosis, it is likely that this is not the only gene causing this disease and probably other genes may be involved in the pathogenesis of the LP disease.
Collapse
Affiliation(s)
- Farzad Izadi
- ENT & HNS Research Center, Hazart Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Frouzandeh Mahjoubi
- Medical Biotechnology Institute, National Institute of Genetic Engineering & Biotechnology (NIGEB), Tehran, Iran
| | - Mohammad Farhadi
- Department & Research Center of Otolaryngology Head & Neck Surgery, Hazrat Rasool Hospital, Tehran University of Medical Sciences & Health Care Services, Tehran, Iran
| | | | | | - Mohammad Moein Tavakoli
- Department & Research Center of Otolaryngology Head & Neck Surgery, Hazrat Rasool Hospital, Tehran University of Medical Sciences & Health Care Services, Tehran, Iran
| | | |
Collapse
|
25
|
Li Y, Li Y, Zhao J, Zheng X, Mao Q, Xia H. Development of a Sensitive Luciferase-Based Sandwich ELISA System for the Detection of Human Extracellular Matrix 1 Protein. Monoclon Antib Immunodiagn Immunother 2016; 35:273-279. [PMID: 27923104 DOI: 10.1089/mab.2016.0033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Enzyme-linked immunosorbent assay (ELISA) has been one of the main methods for detecting an antigen in an aqueous sample for more than four decades. Nowadays, one of the biggest concerns for ELISA is still how to improve the sensitivity of the assay, and the luciferase-luciferin reaction system has been noticed as a new detection method with high sensitivity. In this study, a luciferin-luciferase reaction system was used as the detection method for a sandwich ELISA system. It was shown that this new system led to an increase in the detection sensitivity of at least two times when compared with the traditional horseradish peroxidase (HRP) detection method. Lastly, the serum levels of the human extracellular matrix 1 protein of breast cancer patients were determined by the new system, which were overall similar to the HRP chemiluminescent system. Furthermore, this new luciferase reporter can be implemented into other ELISA systems for the purpose of increasing the assay sensitivity.
Collapse
Affiliation(s)
- Ya Li
- 1 Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University , Xi'an, P.R. China
| | - Yanqing Li
- 1 Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University , Xi'an, P.R. China
| | - Junli Zhao
- 1 Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University , Xi'an, P.R. China
| | - Xiaojing Zheng
- 1 Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University , Xi'an, P.R. China
| | - Qinwen Mao
- 2 Department of Pathology, Northwestern University Feinberg School of Medicine Chicago , Chicago, Illinois
| | - Haibin Xia
- 1 Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University , Xi'an, P.R. China
| |
Collapse
|
26
|
Stålberg P, Westin G, Thirlwell C. Genetics and epigenetics in small intestinal neuroendocrine tumours. J Intern Med 2016; 280:584-594. [PMID: 27306880 DOI: 10.1111/joim.12526] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neuroendocrine tumour of the small intestine (SI-NET), formerly known as midgut carcinoid tumour, is the most common small intestinal malignancy. The incidence is rising, with recent reports of 0.67 per 100 000 in the USA and 1.12 per 100 000 in Sweden. SI-NETs often present a challenge in terms of diagnosis and treatment, as patients often have widespread disease and are beyond cure by surgery. Somatostatin analogues provide the mainstay of medical treatment to control hormonal excess and increase the time to progression. Despite overall favourable prognosis (5-year overall survival of 65%), there is a need to find markers to identify both patients with worse outcome and new targets for therapy. Loss on chromosome 18 has been reported in 60-90% of SI-NETs, but mutated genes on this chromosome have failed detection. Recently, a putative tumour suppressor role has been suggested for TCEB3C occurring at 18q21 (encoding elongin A3), which may undergo epigenetic repression. CDKN1B has recently been revealed as the only recurrently mutated gene in SI-NETs but, with a frequency as low as 8%, its role as a driver in SI-NET development may be questioned. Integrated genomewide analysis including exome and whole-genome sequencing, gene expression, DNA methylation and copy number analysis has identified three novel molecular subtypes of SI-NET with differing clinical outcome. DNA methylation analysis has demonstrated that SI-NETs have significant epigenetic dysregulation in 70-80% of tumours. In this review, we focus on understanding of the genetic, epigenetic and molecular events that lead to development and progression of SI-NETs.
Collapse
Affiliation(s)
- P Stålberg
- Department of Surgical Sciences, Uppsala University and University Hospital, Uppsala, Sweden
| | - G Westin
- Department of Surgical Sciences, Uppsala University and University Hospital, Uppsala, Sweden
| | - C Thirlwell
- Cancer Institute, University College London, London, UK
| |
Collapse
|
27
|
Extracellular matrix 1 (ECM1) regulates the actin cytoskeletal architecture of aggressive breast cancer cells in part via S100A4 and Rho-family GTPases. Clin Exp Metastasis 2016; 34:37-49. [PMID: 27770373 DOI: 10.1007/s10585-016-9827-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 10/12/2016] [Indexed: 10/20/2022]
Abstract
ECM1 overexpression is an independent predictor of poor prognosis in primary breast carcinomas, however the mechanisms by which ECM1 affects tumor progression have not been completely elucidated. ECM1 was silenced in the triple-negative breast cancer cell lines Hs578T and MDAMB231 using siRNA and the cells were evaluated for changes in morphology, migration, invasion and adhesion. Actin cytoskeleton alterations were evaluated by fluorescent staining and levels of activated Rho GTPases by pull down assays. ECM1 downregulation led to significantly diminished cell migration (p = 0.0005 for Hs578T and p = 0.02 for MDAMB231) and cell adhesion (p < 0.001 for Hs578T and p = 0.01 for MDAMB231). Cell invasion (matrigel) was reduced only in the Hs578T cells (p < 0.01). Silencing decreased the expression of the prometastatic molecules S100A4 and TGFβR2 in both cell lines and CD44 in Hs578T cells. ECM1-silenced cells also exhibited alterations in cell shape and showed bundles of F-actin across the cell (stress fibers) whereas NT-siRNA treated cells showed peripheral membrane ruffling. Downregulation of ECM1 was also associated with an increased F/G actin ratio, when compared to the cells transfected with NT siRNA (p < 0.001 for Hs578T and p < 0.00035 for MDAMB231) and a concomitant decline of activated Rho A in the Hs578T cells. Re-expression of S100A4 in ECM1-silenced cells rescued the phenotype in the Hs578T cells but not the MDAMB231 cells. We conclude that ECM1 is a key player in the metastatic process and regulates the actin cytoskeletal architecture of aggressive breast cancer cells at least in part via alterations in S100A4 and Rho A.
Collapse
|
28
|
Thomsen MS, Birkelund S, Burkhart A, Stensballe A, Moos T. Synthesis and deposition of basement membrane proteins by primary brain capillary endothelial cells in a murine model of the blood-brain barrier. J Neurochem 2016; 140:741-754. [PMID: 27456748 DOI: 10.1111/jnc.13747] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 06/25/2016] [Accepted: 07/20/2016] [Indexed: 11/30/2022]
Abstract
The brain vascular basement membrane is important for both blood-brain barrier (BBB) development, stability, and barrier integrity and the contribution hereto from brain capillary endothelial cells (BCECs), pericytes, and astrocytes of the BBB is probably significant. The aim of this study was to analyse four different in vitro models of the murine BBB for expression and possible secretion of major basement membrane proteins from murine BCECs (mBCECs). mBCECs, pericytes and glial cells (mainly astrocytes and microglia) were prepared from brains of C57BL/6 mice. The mBCECs were grown as monoculture, in co-culture with pericytes or mixed glial cells, or as a triple-culture with both pericytes and mixed glial cells. The integrity of the BBB models was validated by measures of transendothelial electrical resistance (TEER) and passive permeability to mannitol. The expression of basement membrane proteins was analysed using RT-qPCR, mass spectrometry and immunocytochemistry. Co-culturing mBCECs with pericytes, mixed glial cells, or both significantly increased the TEER compared to the monoculture, and a low passive permeability was correlated with high TEER. The mBCECs expressed all major basement membrane proteins such as laminin-411, laminin-511, collagen [α1(IV)]2 α2(IV), agrin, perlecan, and nidogen 1 and 2 in vitro. Increased expression of the laminin α5 subunit correlated with the addition of BBB-inducing factors (hydrocortisone, Ro 20-1724, and pCPT-cAMP), whereas increased expression of collagen IV α1 primarily correlated with increased levels of cAMP. In conclusion, BCECs cultured in vitro coherently form a BBB and express basement membrane proteins as a feature of maturation. Cover Image for this issue: doi: 10.1111/jnc.13789.
Collapse
Affiliation(s)
- Maj Schneider Thomsen
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Svend Birkelund
- Laboratory of Medical Mass Spectrometry, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Annette Burkhart
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Allan Stensballe
- Laboratory of Medical Mass Spectrometry, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Torben Moos
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
29
|
Jacobs R, Maasdorp E, Malherbe S, Loxton AG, Stanley K, van der Spuy G, Walzl G, Chegou NN. Diagnostic Potential of Novel Salivary Host Biomarkers as Candidates for the Immunological Diagnosis of Tuberculosis Disease and Monitoring of Tuberculosis Treatment Response. PLoS One 2016; 11:e0160546. [PMID: 27487181 PMCID: PMC4972428 DOI: 10.1371/journal.pone.0160546] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 07/21/2016] [Indexed: 01/05/2023] Open
Abstract
Background There is an urgent need for new tools for the early diagnosis of TB disease and monitoring of the response to treatment, especially in resource-constrained settings. We investigated the usefulness of host markers detected in saliva as candidate biomarkers for the immunological diagnosis of TB disease and monitoring of treatment response. Methods We prospectively collected saliva samples from 51 individuals that presented with signs and symptoms suggestive of TB disease at a health centre in Cape Town, South Africa, prior to the establishment of a clinical diagnosis. Patients were later classified as having TB disease or other respiratory disease (ORD), using a combination of clinical, radiological and laboratory findings. We evaluated the concentrations of 69 host markers in saliva samples using a multiplex cytokine platform, and assessed the diagnostic potentials of these markers by receiver operator characteristics (ROC) curve analysis, and general discriminant analysis. Results Out of the 51 study participants, 18 (35.4%) were diagnosed with TB disease and 12 (23.5%) were HIV infected. Only two of the 69 host markers that were evaluated (IL-16 and IL-23) diagnosed TB disease individually with area under the ROC curve ≥0.70. A five-marker biosignature comprising of IL-1β, IL-23, ECM-1, HCC1 and fibrinogen diagnosed TB disease with a sensitivity of 88.9% (95% CI,76.7–99.9%) and specificity of 89.7% (95% CI, 60.4–96.6%) after leave-one-out cross validation, regardless of HIV infection status. Eight-marker biosignatures performed with a sensitivity of 100% (95% CI, 83.2–100%) and specificity of 95% (95% CI, 68.1–99.9%) in the absence of HIV infection. Furthermore, the concentrations of 11 of the markers changed during treatment, indicating that they may be useful in monitoring of TB treatment response. Conclusion We have identified novel salivary biosignatures which may be useful in the diagnosis of TB disease and monitoring of the response to TB treatment. Our findings require further validation in larger studies before these biosignatures could be considered for point-of-care screening test development.
Collapse
Affiliation(s)
- Ruschca Jacobs
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research and SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Elizna Maasdorp
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research and SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Stephanus Malherbe
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research and SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Andre G. Loxton
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research and SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Kim Stanley
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research and SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gian van der Spuy
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research and SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research and SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Novel N. Chegou
- DST/NRF Centre of Excellence for Biomedical Tuberculosis Research and SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- * E-mail:
| |
Collapse
|
30
|
Chen H, Jia W, Li J. ECM1 promotes migration and invasion of hepatocellular carcinoma by inducing epithelial-mesenchymal transition. World J Surg Oncol 2016; 14:195. [PMID: 27460906 PMCID: PMC4962417 DOI: 10.1186/s12957-016-0952-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/20/2016] [Indexed: 11/25/2022] Open
Abstract
Background Extracellular matrix protein 1 (ECM1) is a glycoprotein involved in many biologic processes. To determine the expression of ECM1 in hepatocellular carcinoma (HCC), and to study the role of ECM1 in inducing epithelia-mesenchymal transition (EMT) to analyze the effect of ECM1 on the migration and invasion of HCC cells. Methods The expression of ECM1 in HCC specimens was examined by immunohistochemistry staining, and the correlations were analyzed between the expression of ECM1 and the clinicopathological data. The ECM1 expression plasmid was transfected into Bel-7402 cells to induce exogenous overexpression of ECM1 protein. The changes of the expression of ECM1, EMT-related protein (E-cadherin, Vimentin), in Bel-7402 cells were detected by Western blot after transfection of ECM1; the wound healing and invasion assay in vitro were used to determine the role of ECM1 gene transfection on the ability of migration and invasive potential of Bel-7402 cells. Results Immumohistochemistry staining method displayed the ECM1 expression was positively associated with vascular invasion, TNM stage, and poor prognosis. A significant positive correlation was found between the expressions of ECM1 and Vimentin. After ECM1 overexpression, Western blot exhibited that the expression of E-cadherin was down-regulated and Vimentin expression was up-regulated in Bel-7402 cells; the wound healing and invasion assay showed that the migration and invasion potentials of Bel-7402 cells were significantly enhanced. Conclusions ECM1, which displayed a high expression in HCC specimens, was closely associated with clinicopathologic data and may promote migration and invasion of HCC cells by inducing EMT. Electronic supplementary material The online version of this article (doi:10.1186/s12957-016-0952-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hao Chen
- Department of Hepatic Surgery, Affiliated Provincial Hospital, Anhui Medical University, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, 17 Lujiang Road, Hefei, 230001, Anhui Province, People's Republic of China
| | - Weidong Jia
- Department of Hepatic Surgery, Affiliated Provincial Hospital, Anhui Medical University, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, 17 Lujiang Road, Hefei, 230001, Anhui Province, People's Republic of China
| | - Jiansheng Li
- Department of Hepatic Surgery, Affiliated Provincial Hospital, Anhui Medical University, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, 17 Lujiang Road, Hefei, 230001, Anhui Province, People's Republic of China.
| |
Collapse
|
31
|
Secretome from resident cardiac stromal cells stimulates proliferation, cardiomyogenesis and angiogenesis of progenitor cells. Int J Cardiol 2016; 221:396-403. [PMID: 27404713 DOI: 10.1016/j.ijcard.2016.06.199] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 06/09/2016] [Accepted: 06/25/2016] [Indexed: 12/20/2022]
Abstract
In the heart, tissue-derived signals play a central role on recruiting/activating stem cell sources to induce cardiac lineage specification for maintenance of tissue homeostasis and repair. Cardiac resident stromal cells (CRSCs) may play a pivotal role in cardiac repair throughout their secretome. Here, we performed the characterization of CRSCs and their secretome by analyzing the composition of their culture-derived extracellular matrix (ECM) and conditioned medium (CM) and by investigating their potential effect on adipose-derived stem cell (ADSC) and progenitor cell behavior. We confirmed that CRSCs are a heterogeneous cell population whose secretome is composed by proteins related to cellular growth, immune response and cardiovascular development and function. We also observed that CRSC secretome was unable to change the behavior of ADSCs, except for proliferation. Additionally, CM from CRSCs demonstrated the potential to drive proliferation and cardiac differentiation of H9c2 cells and also the ability to induce angiogenesis in vitro. Our data suggest that the CRSCs can be a source of important modulating signals for cardiac progenitor cell recruitment/activation.
Collapse
|
32
|
Stenemo M, Teleman J, Sjöström M, Grubb G, Malmström E, Malmström J, Niméus E. Cancer associated proteins in blood plasma: Determining normal variation. Proteomics 2016; 16:1928-37. [PMID: 27121749 DOI: 10.1002/pmic.201500204] [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: 06/02/2015] [Revised: 03/12/2016] [Accepted: 04/15/2016] [Indexed: 11/07/2022]
Abstract
Protein biomarkers have the potential to improve diagnosis, stratification of patients into treatment cohorts, follow disease progression and treatment response. One distinct group of potential biomarkers comprises proteins which have been linked to cancer, known as cancer associated proteins (CAPs). We determined the normal variation of 86 CAPs in 72 individual plasma samples collected from ten individuals using SRM mass spectrometry. Samples were collected weekly during 5 weeks from ten volunteers and over one day at nine fixed time points from three volunteers. We determined the degree of the normal variation depending on interpersonal variation, variation due to time of day, and variation over weeks and observed that the variation dependent on the time of day appeared to be the most important. Subdivision of the proteins resulted in two predominant protein groups containing 21 proteins with relatively high variation in all three factors (day, week and individual), and 22 proteins with relatively low variation in all factors. We present a strategy for prioritizing biomarker candidates for future studies based on stratification over their normal variation and have made all data publicly available. Our findings can be used to improve selection of biomarker candidates in future studies and to determine which proteins are most suitable depending on study design.
Collapse
Affiliation(s)
- Markus Stenemo
- Department of Clinical Sciences Lund, Division of Infection Medicine, Lund University, Lund, Sweden.,Department of Clinical Sciences Lund, Oncology and Pathology, Lund University, Lund, Sweden
| | - Johan Teleman
- Department of Clinical Sciences Lund, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Martin Sjöström
- Department of Clinical Sciences Lund, Oncology and Pathology, Lund University, Lund, Sweden
| | - Gabriel Grubb
- Department of Clinical Sciences Lund, Oncology and Pathology, Lund University, Lund, Sweden
| | - Erik Malmström
- Department of Clinical Sciences Lund, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Johan Malmström
- Department of Clinical Sciences Lund, Division of Infection Medicine, Lund University, Lund, Sweden
| | - Emma Niméus
- Department of Clinical Sciences Lund, Oncology and Pathology, Lund University, Lund, Sweden.,Skåne University Hospital, Department of Surgery, Lund, Sweden
| |
Collapse
|
33
|
Production and characterization of domain-specific monoclonal antibodies against human ECM1. Protein Expr Purif 2016; 121:103-11. [PMID: 26826312 DOI: 10.1016/j.pep.2016.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 12/13/2022]
Abstract
Human extracellular matrix protein-1 (hECM1), a secreted glycoprotein, is widely expressed in different tissues and organs. ECM1 has been implicated in multiple biological functions, which are potentially mediated by the interaction of different ECM1 domains with its ligands. However, the exact biological functions of ECM1 have not been elucidated yet, and the functional study of ECM1 has been partially hampered by the lack of sensitive and specific antibodies, especially those targeting different ECM1 domains. In this study, six strains of monoclonal antibody (MAb) against hECM1 were generated using purified, prokaryotically-expressed hECM1 as an immunogen. The MAbs were shown to be highly sensitive and specific, and suitable for western blot, immunoprecipitation assays and immunohistochemistry. Furthermore, the particular ECM1 domains recognized by different MAbs were identified. Lastly, the MAbs were found to have neutralizing activities, inhibiting the proliferation, migration and metastasis of MDA-MB-231 cells. In conclusion, the domain-specific anti-ECM1 MAbs produced in this study should provide a useful tool for investigating ECM1's biological functions, and cellular pathways in which it is involved.
Collapse
|
34
|
Beretov J, Wasinger VC, Millar EKA, Schwartz P, Graham PH, Li Y. Proteomic Analysis of Urine to Identify Breast Cancer Biomarker Candidates Using a Label-Free LC-MS/MS Approach. PLoS One 2015; 10:e0141876. [PMID: 26544852 PMCID: PMC4636393 DOI: 10.1371/journal.pone.0141876] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 10/14/2015] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Breast cancer is a complex heterogeneous disease and is a leading cause of death in women. Early diagnosis and monitoring progression of breast cancer are important for improving prognosis. The aim of this study was to identify protein biomarkers in urine for early screening detection and monitoring invasive breast cancer progression. METHOD We performed a comparative proteomic analysis using ion count relative quantification label free LC-MS/MS analysis of urine from breast cancer patients (n = 20) and healthy control women (n = 20). RESULTS Unbiased label free LC-MS/MS-based proteomics was used to provide a profile of abundant proteins in the biological system of breast cancer patients. Data analysis revealed 59 urinary proteins that were significantly different in breast cancer patients compared to the normal control subjects (p<0.05, fold change >3). Thirty-six urinary proteins were exclusively found in specific breast cancer stages, with 24 increasing and 12 decreasing in their abundance. Amongst the 59 significant urinary proteins identified, a list of 13 novel up-regulated proteins were revealed that may be used to detect breast cancer. These include stage specific markers associated with pre-invasive breast cancer in the ductal carcinoma in-situ (DCIS) samples (Leucine LRC36, MAST4 and Uncharacterized protein CI131), early invasive breast cancer (DYH8, HBA, PEPA, uncharacterized protein C4orf14 (CD014), filaggrin and MMRN2) and metastatic breast cancer (AGRIN, NEGR1, FIBA and Keratin KIC10). Preliminary validation of 3 potential markers (ECM1, MAST4 and filaggrin) identified was performed in breast cancer cell lines by Western blotting. One potential marker MAST4 was further validated in human breast cancer tissues as well as individual human breast cancer urine samples with immunohistochemistry and Western blotting, respectively. CONCLUSIONS Our results indicate that urine is a useful non-invasive source of biomarkers and the profile patterns (biomarkers) identified, have potential for clinical use in the detection of BC. Validation with a larger independent cohort of patients is required in the following study.
Collapse
Affiliation(s)
- Julia Beretov
- Cancer Care Centre, St George Hospital, Kogarah, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Kensington, Australia
- SEALS, Anatomical Pathology, St George Hospital, Kogarah, Australia
| | - Valerie C. Wasinger
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, UNSW, Kensington, Australia
- School of Medical Sciences, UNSW, Kensington, Australia
| | - Ewan K. A. Millar
- SEALS, Anatomical Pathology, St George Hospital, Kogarah, Australia
- School of Medical Sciences, UNSW, Kensington, Australia
- Cancer Research Program, Kinghorn Cancer Centre and Garvan Institute of Medical Research, Darlinghurst, Australia
- School of Medicine and Health Sciences, University of Western Sydney, Campbelltown, Australia
| | - Peter Schwartz
- Breast Surgery, St George Private Hospital, Kogarah, Australia
| | - Peter H. Graham
- Cancer Care Centre, St George Hospital, Kogarah, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Kensington, Australia
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales (UNSW), Kensington, Australia
- * E-mail:
| |
Collapse
|
35
|
Yabluchanskiy A, Ma Y, DeLeon-Pennell KY, Altara R, Halade GV, Voorhees AP, Nguyen NT, Jin YF, Winniford MD, Hall ME, Han HC, Lindsey ML. Myocardial Infarction Superimposed on Aging: MMP-9 Deletion Promotes M2 Macrophage Polarization. J Gerontol A Biol Sci Med Sci 2015; 71:475-83. [PMID: 25878031 DOI: 10.1093/gerona/glv034] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In this study, we examined the combined effect of aging and myocardial infarction on left ventricular remodeling, focusing on matrix metalloproteinase (MMP)-9-dependent mechanisms. We enrolled 55 C57BL/6J wild type (WT) and 85 MMP-9 Null (Null) mice of both sexes at 11-36 months of age and evaluated their response at Day 7 post-myocardial infarction. Plasma MMP-9 levels positively linked to age in WT mice (r = .46, p = .001). MMP-9 deletion improved survival (76% for WT vs 88% for Null, p = .021). Post-myocardial infarction, there was a progressive increase in left ventricular dilation with age in WT but not in Null mice. By inflammatory gene array analysis, WT mice showed linear age-dependent increases in three different proinflammatory genes (C3, CCl4, and CX3CL1; all p < .05), whereas Null mice showed increases in three proinflammatory genes (CCL5, CCL9, and CXCL4; all p < .05) and seven anti-inflammatory genes (CCL1, CCL6, CCR1, IL11, IL1r2, IL8rb, and Mif; all p < .05). Compared with WT, macrophages isolated from Null left ventricle infarct demonstrated enhanced expression of anti-inflammatory M2 markers CD163, MRC1, TGF-β1, and YM1 (all p < .05), without affecting proinflammatory M1 markers. In conclusion, MMP-9 deletion stimulated anti-inflammatory polarization of macrophages to attenuate left ventricle dysfunction in the aging post-myocardial infarction.
Collapse
Affiliation(s)
- Andriy Yabluchanskiy
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson
| | - Yonggang Ma
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson
| | - Kristine Y DeLeon-Pennell
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson
| | - Raffaele Altara
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson
| | - Ganesh V Halade
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson
| | - Andrew P Voorhees
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Department of Mechanical Engineering and
| | - Nguyen T Nguyen
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Department of Electrical and Computer Engineering, University of Texas at San Antonio
| | - Yu-Fang Jin
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Department of Electrical and Computer Engineering, University of Texas at San Antonio
| | - Michael D Winniford
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Cardiology Division, University of Mississippi Medical Center, Jackson
| | - Michael E Hall
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Cardiology Division, University of Mississippi Medical Center, Jackson
| | - Hai-Chao Han
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Department of Mechanical Engineering and
| | - Merry L Lindsey
- Department of Physiology and Biophysics, San Antonio Cardiovascular Proteomics Center, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson. Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS.
| |
Collapse
|
36
|
ECM1 regulates tumor metastasis and CSC-like property through stabilization of β-catenin. Oncogene 2015; 34:6055-65. [PMID: 25746001 DOI: 10.1038/onc.2015.54] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 01/01/2023]
Abstract
Extracellular Matrix Protein 1 (ECM1) is a marker for tumorigenesis and is correlated with invasiveness and poor prognosis in various types of cancer. However, the functional role of ECM1 in cancer metastasis is unclear. Here, we detected high ECM1 level in breast cancer patient sera that was associated with recurrence of tumor. The modulation of ECM1 expression affected not only cell migration and invasion, but also sphere-forming ability and drug resistance in breast cancer cell lines. In addition, ECM1 regulated the gene expression associated with the epithelial to mesenchymal transition (EMT) progression and cancer stem cell (CSC) maintenance. Interestingly, ECM1 increased β-catenin expression at the post-translational level through induction of MUC1, which was physically associated with β-catenin. Indeed, the association between β-catenin and the MUC1 cytoplasmic tail was increased by ECM1. Furthermore, forced expression of β-catenin altered the gene expression that potentiated EMT progression and CSC phenotype maintenance in the cells. These data provide evidence that ECM1 has an important role in cancer metastasis through β-catenin stabilization.
Collapse
|
37
|
Lee KM, Nam K, Oh S, Lim J, Kim YP, Lee JW, Yu JH, Ahn SH, Kim SB, Noh DY, Lee T, Shin I. Extracellular matrix protein 1 regulates cell proliferation and trastuzumab resistance through activation of epidermal growth factor signaling. Breast Cancer Res 2014; 16:479. [PMID: 25499743 PMCID: PMC4308848 DOI: 10.1186/s13058-014-0479-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 11/12/2014] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Extracellular matrix protein 1 (ECM1) is a secreted glycoprotein with putative functions in cell proliferation, angiogenesis and differentiation. Expression of ECM1 in several types of carcinoma suggests that it may promote tumor development. In this study, we investigated the role of ECM1 in oncogenic cell signaling in breast cancer, and potential mechanisms for its effects. METHODS In order to find out the functional role of ECM1, we used the recombinant human ECM1 and viral transduction systems which stably regulated the expression level of ECM1. We examined the effect of ECM1 on cell proliferation and cell signaling in vitro and in vivo. Moreover, tissues and sera of patients with breast cancer were used to confirm the effect of ECM1. RESULTS ECM1 protein was increased in trastuzumab-resistant (TR) cells, in association with trastuzumab resistance and cell proliferation. Through physical interaction with epidermal growth factor receptor (EGFR), ECM1 potentiated the phosphorylation of EGFR and extracellular signal-regulated kinase upon EGF treatment. Moreover, ECM1-induced galectin-3 cleavage through upregulation of matrix metalloproteinase 9 not only improved mucin 1 expression, but also increased EGFR and human epidermal growth factor receptor 3 protein stability as a secondary signaling. CONCLUSIONS ECM1 has important roles in both cancer development and trastuzumab resistance in breast cancer through activation of EGFR signaling.
Collapse
Affiliation(s)
- Kyung-min Lee
- Department of Life Science, Hanyang University, 222 Wangshimni-ro, Seoul, 133-791, Republic of Korea.
| | - Keesoo Nam
- Department of Life Science, Hanyang University, 222 Wangshimni-ro, Seoul, 133-791, Republic of Korea.
| | - Sunhwa Oh
- Department of Life Science, Hanyang University, 222 Wangshimni-ro, Seoul, 133-791, Republic of Korea.
| | - Juyeon Lim
- Department of Life Science, Hanyang University, 222 Wangshimni-ro, Seoul, 133-791, Republic of Korea.
| | - Young-Pil Kim
- Department of Life Science, Hanyang University, 222 Wangshimni-ro, Seoul, 133-791, Republic of Korea.
| | - Jong Won Lee
- Department of Surgery, College of Medicine, University of Ulsan and Asan Medical Center, 88 Olympic 43-ro, Seoul, 138-736, Republic of Korea.
| | - Jong-Han Yu
- Department of Surgery, College of Medicine, University of Ulsan and Asan Medical Center, 88 Olympic 43-ro, Seoul, 138-736, Republic of Korea.
| | - Sei-Hyun Ahn
- Department of Surgery, College of Medicine, University of Ulsan and Asan Medical Center, 88 Olympic 43-ro, Seoul, 138-736, Republic of Korea.
| | - Sung-Bae Kim
- Department of Oncology, College of Medicine, University of Ulsan and Asan Medical Center, 88 Olympic 43-ro, Seoul, 138-736, Republic of Korea.
| | - Dong-Young Noh
- Cancer Research Institute, Seoul National University College of Medicine, 101 Daehak-ro, Seoul, 110-744, Republic of Korea.
| | - Taehoon Lee
- NOVA Cell Technology, 77 Cheongam-ro, Pohang, 790-784, Republic of Korea.
| | - Incheol Shin
- Department of Life Science, Hanyang University, 222 Wangshimni-ro, Seoul, 133-791, Republic of Korea. .,Natural Science Institute, Hanyang University, 222 Wangshimni-ro, Seoul, 133-791, Republic of Korea.
| |
Collapse
|
38
|
Uematsu S, Goto Y, Suzuki T, Sasazawa Y, Dohmae N, Simizu S. N-Glycosylation of extracellular matrix protein 1 (ECM1) regulates its secretion, which is unrelated to lipoid proteinosis. FEBS Open Bio 2014; 4:879-85. [PMID: 25379385 PMCID: PMC4215116 DOI: 10.1016/j.fob.2014.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/06/2014] [Accepted: 10/06/2014] [Indexed: 11/21/2022] Open
Abstract
Extracellular matrix protein 1 (ECM1) plays roles in extracellular matrix formation. Two ECM1 gene mutations observed in lipoid proteinosis patients suppress its secretion. ECM1 is N-glycosylated at Asn354 and Asn444 residues. N-linked glycan at Asn354 negatively regulated secretion of ECM1.
Extracellular matrix protein 1 (ECM1) is expressed in a wide variety of tissues and plays important roles in extracellular matrix formation. Additionally, ECM1 gene mutations cause lipoid proteinosis (LP), a rare skin condition of genetic origin. However, an effective therapeutic approach of LP is not established. Here, we showed that ECM1 gene mutation observed in LP patients significantly suppresses its secretion. As ECM1 has three putative N-glycosylation sites and most of mutated ECM1 observed in LP patients are defective in these N-glycosylation sites, we investigated the correlation between LP and N-glycosylation of ECM1. We identified that the Asn354 and Asn444 residues in ECM1 were N-glycosylated by mass spectrometry analysis. In addition, an N-linked glycan at Asn354 negatively regulated secretion of ECM1, contrary to LP patient-derived mutants. These results indicate that the defect of N-glycosylation in ECM1 is not involved in the aberration of secretion of LP-derived mutated ECM1.
Collapse
Affiliation(s)
- Shiho Uematsu
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama 223-8522, Japan
| | - Yuki Goto
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama 223-8522, Japan
| | | | - Yukiko Sasazawa
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama 223-8522, Japan
| | - Naoshi Dohmae
- Global Research Cluster, RIKEN, Wako 351-0198, Japan
| | - Siro Simizu
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama 223-8522, Japan
- Corresponding author at: 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan. Tel./fax: +81 45 566 1778.
| |
Collapse
|
39
|
Wu Q, Li X, Yang H, Lu C, You J, Zhang Z. Extracellular matrix protein 1 is correlated to carcinogenesis and lymphatic metastasis of human gastric cancer. World J Surg Oncol 2014; 12:132. [PMID: 24779890 PMCID: PMC4016775 DOI: 10.1186/1477-7819-12-132] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 04/20/2014] [Indexed: 01/26/2023] Open
Abstract
Background Tumor-induced lymphangiogenesis is a crucial step in malignant invasion and metastasis. Extracellular matrix protein 1 (ECM1) was recently reported to play a role in lymphangiogenesis. In the present work, we aimed to evaluate the role of ECM1 in gastric cancer and examined whether aberrant expression of ECM1 increased the tumorigenic and metastatic potential of human gastric cancer. Methods The mRNA and protein expression of ECM1 in gastric cancer specimen and the noncancerous counterparts from 77 patients were detected by real-time PCR and immunohistochemistry staining. Lymphatic microvessel density (LMVD) in the corresponding serial sections was assessed by counting the lymphatic microvessels labelled by D2-40. The correlations between ECM1 expression, LMVD, and the clinicopathological parameters were examined. Results ECM1 protein expression was detected in 70.1% (54/77) of gastric cancer specimen, significantly higher than that in the corresponding counterparts (P <0.01). ECM1 mRNA in tumor specimen was also dramatically amplified. Elevated LMVD and ECM1 were positively correlated (P <0.01). In addition, ECM1 protein expression was also closely associated with depth of tumor invasion and TNM stage (P <0.05, respectively). Conclusions ECM1 expression is aberrant elevated in tumor specimen and is closely related to the tumorigenic and metastatic potential of human gastric cancer. Thus, carrying out the protein examination may be beneficial to predict carcinogenesis and metastatic spread of human gastric cancer.
Collapse
Affiliation(s)
| | | | | | | | - Jun You
- Xiamen Cancer Centre, the First Affiliated Hospital of Xiamen University (the Teaching Hospital of Fujian Medical University), Xiamen, China.
| | | |
Collapse
|
40
|
Nikolajsen CL, Dyrlund TF, Poulsen ET, Enghild JJ, Scavenius C. Coagulation factor XIIIa substrates in human plasma: identification and incorporation into the clot. J Biol Chem 2014; 289:6526-6534. [PMID: 24443567 DOI: 10.1074/jbc.m113.517904] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Coagulation factor XIII (FXIII) is a transglutaminase with a well defined role in the final stages of blood coagulation. Active FXIII (FXIIIa) catalyzes the formation of ε-(γ-glutamyl)lysine isopeptide bonds between specific Gln and Lys residues. The primary physiological outcome of this catalytic activity is stabilization of the fibrin clot during coagulation. The stabilization is achieved through the introduction of cross-links between fibrin monomers and through cross-linking of proteins with anti-fibrinolytic activity to fibrin. FXIIIa additionally cross-links several proteins with other functionalities to the clot. Cross-linking of proteins to the clot is generally believed to modify clot characteristics such as proteolytic susceptibility and hereby affect the outcome of tissue damage. In the present study, we use a proteomic approach in combination with transglutaminase-specific labeling to identify FXIIIa plasma protein substrates and their reactive residues. The results revealed a total of 147 FXIIIa substrates, of which 132 have not previously been described. We confirm that 48 of the FXIIIa substrates were indeed incorporated into the insoluble fibrin clot during the coagulation of plasma. The identified substrates are involved in, among other activities, complement activation, coagulation, inflammatory and immune responses, and extracellular matrix organization.
Collapse
Affiliation(s)
- Camilla Lund Nikolajsen
- Department of Molecular Biology and Genetics, Science Park, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Thomas F Dyrlund
- Department of Molecular Biology and Genetics, Science Park, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Ebbe Toftgaard Poulsen
- Department of Molecular Biology and Genetics, Science Park, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Science Park, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark.
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Science Park, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark.
| |
Collapse
|
41
|
Large-scale investigation of Leishmania interaction networks with host extracellular matrix by surface plasmon resonance imaging. Infect Immun 2013; 82:594-606. [PMID: 24478075 DOI: 10.1128/iai.01146-13] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
We have set up an assay to study the interactions of live pathogens with their hosts by using protein and glycosaminoglycan arrays probed by surface plasmon resonance imaging. We have used this assay to characterize the interactions of Leishmania promastigotes with ~70 mammalian host biomolecules (extracellular proteins, glycosaminoglycans, growth factors, cell surface receptors). We have identified, in total, 27 new partners (23 proteins, 4 glycosaminoglycans) of procyclic promastigotes of six Leishmania species and 18 partners (15 proteins, 3 glycosaminoglycans) of three species of stationary-phase promastigotes for all the strains tested. The diversity of the interaction repertoires of Leishmania parasites reflects their dynamic and complex interplay with their mammalian hosts, which depends mostly on the species and strains of Leishmania. Stationary-phase Leishmania parasites target extracellular matrix proteins and glycosaminoglycans, which are highly connected in the extracellular interaction network. Heparin and heparan sulfate bind to most Leishmania strains tested, and 6-O-sulfate groups play a crucial role in these interactions. Numerous Leishmania strains bind to tropoelastin, and some strains are even able to degrade it. Several strains interact with collagen VI, which is expressed by macrophages. Most Leishmania promastigotes interact with several regulators of angiogenesis, including antiangiogenic factors (endostatin, anastellin) and proangiogenic factors (ECM-1, VEGF, and TEM8 [also known as anthrax toxin receptor 1]), which are regulated by hypoxia. Since hypoxia modulates the infection of macrophages by the parasites, these interactions might influence the infection of host cells by Leishmania.
Collapse
|
42
|
Zhu B, Xu T, Yuan J, Guo X, Liu D. Transcriptome sequencing reveals differences between primary and secondary hair follicle-derived dermal papilla cells of the Cashmere goat (Capra hircus). PLoS One 2013; 8:e76282. [PMID: 24069460 PMCID: PMC3777969 DOI: 10.1371/journal.pone.0076282] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 08/22/2013] [Indexed: 12/30/2022] Open
Abstract
The dermal papilla is thought to establish the character and control the size of hair follicles. Inner Mongolia Cashmere goats (Capra hircus) have a double coat comprising the primary and secondary hair follicles, which have dramatically different sizes and textures. The Cashmere goat is rapidly becoming a potent model for hair follicle morphogenesis research. In this study, we established two dermal papilla cell lines during the anagen phase of the hair growth cycle from the primary and secondary hair follicles and clarified the similarities and differences in their morphology and growth characteristics. High-throughput transcriptome sequencing was used to identify gene expression differences between the two dermal papilla cell lines. Many of the differentially expressed genes are involved in vascularization, ECM-receptor interaction and Wnt/β-catenin/Lef1 signaling pathways, which intimately associated with hair follicle morphogenesis. These findings provide valuable information for research on postnatal morphogenesis of hair follicles.
Collapse
Affiliation(s)
- Bing Zhu
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Teng Xu
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Jianlong Yuan
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Xudong Guo
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
- * E-mail: (XG); (DL)
| | - Dongjun Liu
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
- * E-mail: (XG); (DL)
| |
Collapse
|
43
|
Human Melanoma cells over-express extracellular matrix 1 (ECM1) which is regulated by TFAP2C. PLoS One 2013; 8:e73953. [PMID: 24023917 PMCID: PMC3759440 DOI: 10.1371/journal.pone.0073953] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 07/25/2013] [Indexed: 11/19/2022] Open
Abstract
Extracellular matrix 1 (ECM1) is over-expressed in multiple epithelial malignancies. However, knowledge regarding the expression of ECM1 in melanomas and the mechanisms of ECM1 regulation is limited. In this study, we found that ECM1 is over-expressed in several melanoma cell lines, when compared to primary melanocytes, and furthermore, that ECM1 expression paralleled that of TFAP2C levels in multiple cell lines. Knockdown of TFAP2C in the A375 cell line with siRNA led to a reduction in ECM1 expression, and upregulation of TFAP2C with adenoviral vectors in the WM793 cell line resulted in ECM1 upregulation. Utilizing 5’ RACE to identify transcription start sites (TSS) and luciferase reporter assays in the ECM1-overexpressing A375 cell line, we identified the minimal promoter region of human ECM1 and demonstrate that an approximately 100bp fragment upstream of the TSS containing a TATA box and binding sites for AP1, SP1 and Ets is sufficient for promoter activity. Chromatin immunoprecipitation and direct sequencing (ChIP-seq) for TFAP2C in the A375 cell line identified an AP2 regulatory region in the promoter of the ECM1 gene. Gelshift assays further confirmed binding of TFAP2C to this site. ECM1 knockdown reduces melanoma cell attachment and is consistent with findings that ECM1 overexpression has been associated with a poor prognosis. Our investigations show an as yet unrecognized role for TFAP2C in melanoma via its regulation of ECM1.
Collapse
|
44
|
Differential expression of genes associated with the progression of renal disease in the kidneys of liver-specific glucokinase gene knockout mice. Int J Mol Sci 2013; 14:6467-86. [PMID: 23519111 PMCID: PMC3634482 DOI: 10.3390/ijms14036467] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 03/18/2013] [Accepted: 03/19/2013] [Indexed: 12/30/2022] Open
Abstract
Liver glucokinase (GCK) deficient mice possess mild renal complications associated with diabetes. To investigate the progression of kidney disease and identify candidate genes involved in the pathogenesis of renal damage, we examined changes in tissue structure and gene expression in the kidneys of liver-specific GCK knockout (gckw/-) mice and age-matched normal wild-type control (gckw/w) mice as they aged. Suppression subtractive hybridization (SSH) was used to identify candidate genes that showed a pattern of differential expression between kidneys of gckw/- and gckw/w mice at 60 weeks of age. Differential expression of the candidate genes was examined by real-time qPCR in liver-specific gckw/- and gckw/w mice at 16, 26, 40, 60, and 85 weeks of age. Among the candidate genes, only glutathione peroxidase-3 (GPX3) was confirmed to show differential expression by qPCR in the 60-week old mice, however two others genes, MALAT1 and KEG, showed significant changes at other ages. This study shows that liver-specific glucokinase deficient mice display changes in kidney morphology by 40 weeks of age, and that renal complication may be correlated with a reduction in GPX3 levels. Since decreased GPX3 mRNA expression was observed at 26 weeks, which is younger than the age when pathological changes can be seen in kidney biopsies, GPX3 may serve as an early marker for kidney damage.
Collapse
|
45
|
Abstract
UNLABELLED Identification of common molecular mechanisms is needed to facilitate the development of new treatment options for patients with ileal carcinoids. PURPOSE OF REVIEW Recent profiling studies on ileal carcinoids were examined to obtain a comprehensive view of risk factors, genetic aberrations, and transcriptional alterations. Special attention was paid to mechanisms that could provide novel targets for therapy. RESULTS Genome-wide association studies have shown that single nucleotide polymorphisms (SNPs) at IL12A and DAD1 are associated with an increased risk of ileal carcinoids. Genomic profiling revealed distinct patterns of copy-number alterations in ileal carcinoids. Two groups of carcinoids could be identified by hierarchical clustering. A major group of tumors was characterized by loss on chromosome 18 followed by additional losses on chromosomes 3p, 11q, and 13. Three minimal common regions of deletions were identified at 18q21.1-q21.31, 18q22.1-q22.2, and 18q22.3-q23. A minor group of tumors was characterized by clustered gains on chromosomes 4, 5, 7, 14, and 20. Expression profiling identified three groups of ileal carcinoids by principal component analysis. Tumor progression was associated with changes in gene expression including downregulation of MIR133A. Candidate genes for targeted therapy included ERBB2/HER2, DAD1, PRKCA, RYBP, CASP1, CASP4, CASP5, VMAT1, RET, APLP1, OR51E1, GPR112, SPOCK1, RUNX1, and MIR133A. CONCLUSION Profiling of ileal carcinoids has revealed recurrent genetic alterations and distinct patterns of gene expression. Frequent alterations in cellular pathways and genes were identified, suggesting novel targets for therapy. Translational studies are needed to validate suggested molecular targets.
Collapse
Affiliation(s)
- Ola Nilsson
- Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Göteborg, Sweden.
| |
Collapse
|
46
|
Izadi F, Mahjoubi F, Farhadi M, Tavakoli MM, Samanian S. A novel missense mutation in exon 7 of the ECM1 gene in an Iranian lipoid proteinosis patient. GENETICS AND MOLECULAR RESEARCH 2012; 11:3955-60. [PMID: 23212332 DOI: 10.4238/2012.november.14.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Lipoid proteinosis (LP) is a rare autosomal recessive disorder. Classical clinical features include warty skin infiltration, papules on the eyelids, skin scarring, as well as extracutaneous abnormalities such as hoarseness of the voice, epilepsy, and neuropsychiatric abnormalities. A defect in the ECM1 gene is responsible for this disease. A 21-year-old female patient from consanguineous parents (first cousins) was referred to our clinic with many symptoms of LP, such as hoarse voice from infancy, diffuse acneiform scars on her face, and hyperkeratosis on her knees and elbows. The entire ECM1 gene was screened using PCR and sequencing. A novel missense mutation was found in exon 7 of this patient. We report a novel missense mutation in exon 7 of the ECM1 gene found in an Iranian LP patient that causes a C269Y amino acid exchange.
Collapse
Affiliation(s)
- F Izadi
- ENT-Head and Neck Research Center and Department, Hazrat Rasoul Akram Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | |
Collapse
|
47
|
Wu QW, She HQ, Liang J, Huang YF, Yang QM, Yang QL, Zhang ZM. Expression and clinical significance of extracellular matrix protein 1 and vascular endothelial growth factor-C in lymphatic metastasis of human breast cancer. BMC Cancer 2012; 12:47. [PMID: 22284579 PMCID: PMC3292501 DOI: 10.1186/1471-2407-12-47] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 01/27/2012] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Extracellular matrix protein 1 (ECM1) and vascular endothelial growth factor-C (VEGF-C) are secretory glycoproteins that are associated with lymphangiogenesis; these proteins could, therefore, play important roles in the lymphatic dissemination of tumors. However, very little is known about their potential roles in lymphangiogenesis. The aim of this study was to investigate whether correlations exist between ECM1 and VEGF-C in human breast cancer, lymphangiogenesis, and the clinicopathological characteristics of the disease. METHODS ECM1 and VEGF-C mRNA and protein expression levels in 41 patients were investigated using real-time reverse transcriptase polymerase chain reaction (RT-PCR), or immunohistochemical (IHC) staining of breast cancer tissue, matched noncancerous breast epithelial tissues, and suspicious metastatic axillary lymph nodes. D2-40 labelled lymph vessels and lymphatic microvessel density (LMVD) were counted. Correlations between ECM1 or VEGF-C protein expression levels, LMVD, and clinicopathological parameters were statistically tested. RESULTS The rate of ECM1 positive staining in breast cancer tissues was higher (31/41, 75.6%) than that in the corresponding epithelial tissues (4/41, 9.8%, P < 0.001) and lymph nodes (13/41, 31.7%, P < 0.001). Similarly, the VEGF-C expression rate in cancer specimens was higher (33/41, 80.5%) than in epithelial tissues (19/41, 46.3%, P < 0.01) or lymph nodes (15/41, 36.6%, P < 0.01). Higher ECM1 and VEGF-C mRNA expression levels were also detected in the tumor tissues, compared to the non-cancerous tissue types or lymph nodes (P < 0.05). ECM1 protein expression was positively correlated with the estrogen receptor status (P < 0.05) and LMVD (P < 0.05). LMVD in the ECM1- and VEGF-C-positive tumor specimens was higher than that in the tissue types with negative staining (P < 0.05). CONCLUSIONS Both ECM1 and VEGF-C were overexpressed in breast cancer tissue samples. ECM1 expression was positively correlated with estrogen responsiveness and the metastatic properties of breast cancer. We conclude, therefore, that ECM1 and VEGF-C may have a synergistic effect on lymphangiogenesis to facilitate lymphatic metastasis of breast cancer.
Collapse
Affiliation(s)
- Qiu-Wan Wu
- Fujian Medical University, Fuzhou 350108, China
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Hong-Qiang She
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Jing Liang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Yu-Fan Huang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Qing-Mo Yang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Qiao-Lu Yang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Zhi-Ming Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| |
Collapse
|
48
|
Molecular analysis of lipoid proteinosis: identification of a novel nonsense mutation in the ECM1 gene in a Pakistani family. Diagn Pathol 2011; 6:69. [PMID: 21791056 PMCID: PMC3158550 DOI: 10.1186/1746-1596-6-69] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 07/26/2011] [Indexed: 11/17/2022] Open
Abstract
Lipoid proteinosis is a rare autosomal recessive disease characterized by cutaneous and mucosal lesions and hoarseness appearing in early childhood that is caused by homozygous or compound heterozygous mutations in the ECM1 gene located on chromosome 1q21. The aim of the study was to investigate the molecular genetic defect underlying lipoid proteinosis in a consanguineous Pakistani family.
Collapse
|
49
|
López-Marure R, Contreras PG, Dillon JS. Effects of dehydroepiandrosterone on proliferation, migration, and death of breast cancer cells. Eur J Pharmacol 2011; 660:268-74. [PMID: 21497598 DOI: 10.1016/j.ejphar.2011.03.040] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 03/07/2011] [Accepted: 03/28/2011] [Indexed: 12/22/2022]
Abstract
Cancer invasion and metastasis are the leading causes of mortality in patients with breast cancer. Dehydroepiandrosterone (DHEA) has a protective role against cancer, however, the mechanism by which DHEA has this effect remains poorly understood. The present study was aimed at investigating the actions of DHEA on the proliferation, cell cycle, death and migration of breast cancer cell lines. We used MCF-7 cells (estrogen receptors positive) and MDA-MB-231 and Hs578T cells (estrogen receptors negative) for these studies. Cell proliferation was evaluated by crystal violet staining, cell cycle by flow cytometry, and cell death by the carboxyfluorescein FLICA analysis of caspase activation. Migration was evaluated by transwell cell migration and wound assay. We also determined the expression of ECM-1 protein by western blotting and RT-PCR in real time. DHEA inhibited the proliferation of all breast cancer cell lines. This was associated with an arrest in the G1 phase of the cell cycle and death in MCF-7 cells. There was no alteration in any of the cell cycle phases or death in DHEA treated MDA-MB-231 or Hs578T cells. DHEA also suppressed the migration of all breast cancer cell lines, independently of the presence of estrogen receptors and decreased the expression of ECM-1 protein in Hs578T cells. These results suggest that the mechanism of DHEA actions against breast cancer involves the inhibition of cell proliferation and the suppression of migration, indicating that DHEA could be useful in the treatment of breast cancer.
Collapse
Affiliation(s)
- Rebeca López-Marure
- Departamento de Biología Celular, Instituto Nacional de Cardiología Ignacio Chávez, Mexico.
| | | | | |
Collapse
|
50
|
Li Z, Zhang Y, Liu Z, Wu X, Zheng Y, Tao Z, Mao K, Wang J, Lin G, Tian L, Ji Y, Qin M, Sun S, Zhu X, Sun B. ECM1 controls T(H)2 cell egress from lymph nodes through re-expression of S1P(1). Nat Immunol 2011; 12:178-85. [PMID: 21217760 DOI: 10.1038/ni.1983] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 12/08/2010] [Indexed: 12/13/2022]
Abstract
Type 2 helper T cells (T(H)2) are critically involved in allergies and asthma. Here we demonstrate that extracellular matrix protein-1 (ECM1) is highly and selectively expressed in T(H)2 cells. ECM1 deficiency caused impaired T(H)2 responses and reduced allergic airway inflammation in vivo. Functional analysis demonstrated that although the T(H)2 polarization of ECM1-deficient cells was unimpaired, these cells had a defect in migration and were retained in peripheral lymphoid organs. This was associated with reduced expression of KLF2 and S1P(1). We also found that ECM1 could directly bind the interleukin-2 (IL-2) receptor to inhibit IL-2 signaling and activate S1P(1) expression. Our data identify a previously unknown function of ECM1 in regulating T(H)2 cell migration through control of KLF2 and S1P(1) expression.
Collapse
Affiliation(s)
- Zhenhu Li
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|