1
|
Files R, Cardoso C, Prada J, Silva F, Pires I. Syndecan-1 and E-Cadherin Expression in Canine Cutaneous Squamous Cell Carcinoma. Vet Sci 2024; 11:652. [PMID: 39728992 DOI: 10.3390/vetsci11120652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) in dogs is a locally invasive tumor that typically occurs in areas of poorly pigmented skin due to sun exposure. Identifying new biomarkers, such as syndecan-1 (CD138) and E-cadherin, is fundamental for tumor diagnosis and prognosis. Dysregulation of syndecan-1, expressed in epithelial tissue, fibroblasts, and plasma cells, is associated with poor prognosis in several types of cancer. Similarly, E-cadherin, which plays a crucial role in cell adhesion and epithelial functionality, is also linked to adverse outcomes. This study evaluated the expression of syndecan-1 and E-cadherin in 47 cases of canine cutaneous squamous cell carcinoma. The results showed that the intensity of syndecan-1 decreased with increasing tumor aggressiveness, and its presence in the stroma was significantly associated with tumor grade. E-cadherin also demonstrated a decrease in intensity with increasing malignancy. However, the association between syndecan-1 and E-cadherin was not statistically significant. E-cadherin reduction and stromal syndecan-1 positivity seem to be associated with tumor aggressiveness in canine cutaneous squamous cell carcinoma. Further studies are needed to explore their roles in tumor progression.
Collapse
Affiliation(s)
- Rita Files
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Cláudia Cardoso
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Justina Prada
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Animal and Veterinary Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Filipe Silva
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Animal and Veterinary Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| | - Isabel Pires
- Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
- Animal and Veterinary Research Centre (CECAV), Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), University of Trás-os-Montes and Alto Douro, 5000-801 Vila Real, Portugal
| |
Collapse
|
2
|
Sadique FL, Subramaiam H, Krishnappa P, Chellappan DK, Ma JH. Recent advances in breast cancer metastasis with special emphasis on metastasis to the brain. Pathol Res Pract 2024; 260:155378. [PMID: 38850880 DOI: 10.1016/j.prp.2024.155378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Understanding the underlying mechanisms of breast cancer metastasis is of vital importance for developing treatment approaches. This review emphasizes contemporary breakthrough studies with special focus on breast cancer brain metastasis. Acquired mutational changes in metastatic lesions are often distinct from the primary tumor, suggesting altered mutagenesis pathways. The concept of micrometastases and heterogeneity within the tumors unravels novel therapeutic targets at genomic and molecular levels through epigenetic and proteomic profiling. Several pre-clinical studies have identified mechanisms involving the immune system, where tumor associated macrophages are key players. Expression of cell proteins like Syndecan1, fatty acid-binding protein 7 and tropomyosin kinase receptor B have been implicated in aiding the transmigration of breast cancer cells to the brain. Changes in the proteomic landscape of the blood-brain-barrier show altered permeability characteristics, supporting entry of cancer cells. Findings from laboratory studies pave the path for the emergence of new biomarkers, especially blood-based miRNA and circulating tumor cell markers for prognostic staging. The constantly evolving therapeutics call for clinical trials backing supportive evidence of efficacies of both novel and existing approaches. The challenge lying ahead is discovering innovative techniques to replace use of human samples and optimize small-scale patient recruitment in trials.
Collapse
Affiliation(s)
- Fairooz Labiba Sadique
- Department of Biomedical Science, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Hemavathy Subramaiam
- Division of Pathology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia.
| | - Purushotham Krishnappa
- Division of Pathology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Jin Hao Ma
- School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
3
|
Rajabloo Y, Saberi-Karimian M, Soflaei SS, Ferns GA, Ghayour-Mobarhan M. Syndecans and diabetic complications: A narrative review. Am J Med Sci 2024; 368:99-111. [PMID: 38697476 DOI: 10.1016/j.amjms.2024.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 04/20/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024]
Abstract
Syndecan (SDC) is a member of the heparan sulfate proteoglycan (HSPG) family. It appears to play a role in the aetiology of diabetic complications, with decreased levels of SDCs being reported in the kidney, retina, and cardiac muscle in models of diabetes mellitus (DM). The reduced levels of SDCs may play an important role in the development of albuminuria in DM. Some studies have provided the evidence supporting the mechanisms underlying the role of SDCs in DM. However, SDCs and the molecular mechanisms involved are complex and need to be further elucidated. This review focuses on the underlying molecular mechanisms of SDCs that are involved in the development and progression of the complications of DM, which may help in developing new strategies to prevent and treat these complications.
Collapse
Affiliation(s)
- Yasamin Rajabloo
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Saberi-Karimian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran; Endoscopic and Minimally Invasive Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Sara Saffar Soflaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Majid Ghayour-Mobarhan
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Rodriguez-Jimenez NA, Gonzalez-Ponce F, Gamez-Nava JI, Ramirez-Villafaña M, Saldaña-Cruz AM, Ponce-Guarneros JM, Olivas-Flores EM, Macías-Islas MA, Valdivia-Tangarife ER, Jacobo-Cuevas H, Ramos-Estrada LG, Totsuka-Sutto S, Cardona-Muñoz EG, Gonzalez-Lopez L. Syndecan-1 Levels in Females with Active Rheumatoid Arthritis. J Clin Med 2024; 13:4110. [PMID: 39064151 PMCID: PMC11278313 DOI: 10.3390/jcm13144110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Background: The relationship between serum glycoprotein syndecan-1 and disease activity in rheumatoid arthritis (RA) is still unknown. This study aimed to evaluate whether serum syndecan-1 concentrations are associated with moderate/severe disease activity. Methods: Study Design: This was a cross-sectional study. Seventy-five adult women with RA were classified into (a) moderate/severe RA based on the disease activity score, using the erythrocyte sedimentation rate (DAS28-ESR ≥ 3.2, n = 50), and (b) RA in remission (DAS28-ESR < 2.6, n = 25). Twenty-five healthy women were taken as the reference group. Syndecan-1 levels were determined using enzyme-linked immunosorbent assay (ELISA). High values of serum syndecan-1 levels (≥24 ng/mL) were used to identify the utility values of this biomarker. Results: The patients with RA had higher levels of syndecan-1 than the controls (p < 0.001). RA patients with active disease had higher syndecan-1 levels than RA patients in remission (57.6 vs. 23.5 ng/mL, respectively; p = 0.002). High syndecan-1 concentrations demonstrated the following utility values for identifying disease activity: sensitivity, 84% (95%CI: 71-93); specificity, 52% (95%CI: 31-72); positive predictive value, 78% (95%CI: 70-84); and negative predictive value, 62% (95%CI: 44-77). Conclusions: High syndecan-1 levels have good sensitivity and positive predictive value for identifying disease activity; however, their specificity is limited. Future prospective studies are needed to assess whether syndecan-1 levels can predict treatment failure in RA.
Collapse
Affiliation(s)
- Norma Alejandra Rodriguez-Jimenez
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.A.R.-J.); (F.G.-P.); (J.I.G.-N.); (M.R.-V.); (A.M.S.-C.); (J.M.P.-G.); (L.G.R.-E.); (S.T.-S.); (E.G.C.-M.)
| | - Fabiola Gonzalez-Ponce
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.A.R.-J.); (F.G.-P.); (J.I.G.-N.); (M.R.-V.); (A.M.S.-C.); (J.M.P.-G.); (L.G.R.-E.); (S.T.-S.); (E.G.C.-M.)
| | - Jorge Ivan Gamez-Nava
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.A.R.-J.); (F.G.-P.); (J.I.G.-N.); (M.R.-V.); (A.M.S.-C.); (J.M.P.-G.); (L.G.R.-E.); (S.T.-S.); (E.G.C.-M.)
- Programa de Maestría en Salud Publica, Departamento de Salud Publica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Melissa Ramirez-Villafaña
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.A.R.-J.); (F.G.-P.); (J.I.G.-N.); (M.R.-V.); (A.M.S.-C.); (J.M.P.-G.); (L.G.R.-E.); (S.T.-S.); (E.G.C.-M.)
| | - Ana Miriam Saldaña-Cruz
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.A.R.-J.); (F.G.-P.); (J.I.G.-N.); (M.R.-V.); (A.M.S.-C.); (J.M.P.-G.); (L.G.R.-E.); (S.T.-S.); (E.G.C.-M.)
| | - Juan Manuel Ponce-Guarneros
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.A.R.-J.); (F.G.-P.); (J.I.G.-N.); (M.R.-V.); (A.M.S.-C.); (J.M.P.-G.); (L.G.R.-E.); (S.T.-S.); (E.G.C.-M.)
- Unidad Medica Familiar 97, Instituto Mexicano del Seguro Social, Magdalena 46470, Jalisco, Mexico
| | - Eva Maria Olivas-Flores
- Departamento de Anestesiología, Hospital de Especialidades, Centro Medico Nacional de Occidente, IMSS, Guadalajara 44340, Jalisco, Mexico;
| | - Miguel Angel Macías-Islas
- Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.A.M.-I.); (E.R.V.-T.)
| | - Edgar Ricardo Valdivia-Tangarife
- Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (M.A.M.-I.); (E.R.V.-T.)
| | - Heriberto Jacobo-Cuevas
- Programa de Postdoctorado en el Departamento de Psicología Básica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
| | - Luz Gabriela Ramos-Estrada
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.A.R.-J.); (F.G.-P.); (J.I.G.-N.); (M.R.-V.); (A.M.S.-C.); (J.M.P.-G.); (L.G.R.-E.); (S.T.-S.); (E.G.C.-M.)
| | - Sylvia Totsuka-Sutto
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.A.R.-J.); (F.G.-P.); (J.I.G.-N.); (M.R.-V.); (A.M.S.-C.); (J.M.P.-G.); (L.G.R.-E.); (S.T.-S.); (E.G.C.-M.)
| | - Ernesto German Cardona-Muñoz
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.A.R.-J.); (F.G.-P.); (J.I.G.-N.); (M.R.-V.); (A.M.S.-C.); (J.M.P.-G.); (L.G.R.-E.); (S.T.-S.); (E.G.C.-M.)
| | - Laura Gonzalez-Lopez
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (N.A.R.-J.); (F.G.-P.); (J.I.G.-N.); (M.R.-V.); (A.M.S.-C.); (J.M.P.-G.); (L.G.R.-E.); (S.T.-S.); (E.G.C.-M.)
- Programa de Maestría en Salud Publica, Departamento de Salud Publica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | | |
Collapse
|
5
|
Antwi FD, Awad T, Larin M, Heesom K, Lewis P, Reddell P, Poghosyan Z, Dewitt S, Moseley R, Knäuper V. Tigilanol Tiglate-Induced Changes in Secretome Profiles Alter C-Met Phosphorylation and Cell Surface Protein Expression in H357 Head and Neck Cancer Cells. Cells 2024; 13:982. [PMID: 38891113 PMCID: PMC11171882 DOI: 10.3390/cells13110982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Tigilanol tiglate (TT, also known as EBC-46) is a novel, plant-derived diterpene ester possessing anticancer and wound-healing properties. Here, we show that TT-evoked PKC-dependent S985 phosphorylation of the tyrosine kinase MET leads to subsequent degradation of tyrosine phosphorylated p-Y1003 and p-Y1234/5 MET species. PKC inhibition with BIM-1 blocked S985 phosphorylation of MET and led to MET cell surface accumulation. Treatment with metalloproteinase inhibitors prevented MET-ECD release into cell culture media, which was also blocked by PKC inhibitors. Furthermore, unbiased secretome analysis, performed using TMT-technology, identified additional targets of TT-dependent release of cell surface proteins from H357 head and neck cancer cells. We confirm that the MET co-signalling receptor syndecan-1 was cleaved from the cell surface in response to TT treatment. This was accompanied by rapid cleavage of the cellular junction adhesion protein Nectin-1 and the nerve growth factor receptor NGFRp75/TNFR16. These findings, that TT is a novel negative regulator of protumorigenic c-MET and NGFRp75/TNFR16 signalling, as well as regulating Nectin-1-mediated cell adhesion, further contribute to our understanding of the mode of action and efficacy of TT in the treatment of solid tumours.
Collapse
Affiliation(s)
- Frank Dickson Antwi
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK (S.D.); (R.M.)
| | - Tufaha Awad
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK (S.D.); (R.M.)
| | - Meghan Larin
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK (S.D.); (R.M.)
| | - Kate Heesom
- Bristol Proteomics Facility, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | - Phil Lewis
- Bristol Proteomics Facility, Biomedical Sciences Building, University Walk, University of Bristol, Bristol BS8 1TD, UK
| | | | - Zaruhi Poghosyan
- School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XN, UK
| | - Sharon Dewitt
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK (S.D.); (R.M.)
| | - Ryan Moseley
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK (S.D.); (R.M.)
| | - Vera Knäuper
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, UK (S.D.); (R.M.)
| |
Collapse
|
6
|
Shen K, Chen B, Yang L, Gao W. KYNU as a Biomarker of Tumor-Associated Macrophages and Correlates with Immunosuppressive Microenvironment and Poor Prognosis in Gastric Cancer. Int J Genomics 2023; 2023:4662480. [PMID: 37954130 PMCID: PMC10635752 DOI: 10.1155/2023/4662480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/14/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023] Open
Abstract
Background Kynureninase (KYNU) is a potential prognostic marker for various tumor types. However, no reports on the biological effects and prognostic value of KYNU in gastric cancer (GC) exist. Methods GC-associated single-cell RNA sequencing and bulk RNA sequencing (bulk-seq) data were obtained from the Gene Expression Omnibus and The Cancer Genome Atlas databases, respectively. The differential expression of KYNU between GC and normal gastric tissues was first analyzed based on the bulk-seq data, followed by an exploration of the relationship between KYNU and various clinicopathological features. The Kaplan-Meier survival and Cox regression analyses were performed to determine the prognostic value of KYNU. The relationship between KYNU expression and immune cell infiltration and immune checkpoints was also explored. The biological function of KYNU was further examined at the single-cell level, and in vitro experiments were performed to examine the effect of KYNU on GC cell proliferation and invasion. Results KYNU expression was significantly elevated in GC samples. Clinical features and survival analysis indicated that high KYNU expression was associated with poor clinical phenotypes and prognosis, whereas Cox analysis showed that KYNU was an independent risk factor for patients with GC. Notably, high expression of KYNU induced a poor immune microenvironment and contributed to the upregulation of immune checkpoints. KYNU-overexpressing macrophages drove GC progression through unique ligand-receptor pairs and transcription factors and were associated with adverse clinical phenotypes in GC. KYNU was overexpressed in GC cells in vitro, and KYNU knockout significantly inhibited GC cell proliferation and invasion. Conclusion High KYNU expression promotes an adverse immune microenvironment and low survival rates in GC. KYNU and KYNU-related macrophages may serve as novel molecular targets in the treatment of GC.
Collapse
Affiliation(s)
- Kaiyu Shen
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Binyu Chen
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Liu Yang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wencang Gao
- Department of Oncology, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310005, China
| |
Collapse
|
7
|
Kaur G, Harris NR. Endothelial glycocalyx in retina, hyperglycemia, and diabetic retinopathy. Am J Physiol Cell Physiol 2023; 324:C1061-C1077. [PMID: 36939202 PMCID: PMC10125029 DOI: 10.1152/ajpcell.00188.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 02/28/2023] [Accepted: 03/16/2023] [Indexed: 03/21/2023]
Abstract
The endothelial glycocalyx (EG) is a meshlike network present on the apical surface of the endothelium. Membrane-bound proteoglycans, the major backbone molecules of the EG, consist of glycosaminoglycans attached to core proteins. In addition to maintaining the integrity of the endothelial barrier, the EG regulates inflammation and perfusion and acts as a mechanosensor. The loss of the EG can cause endothelial dysfunction and drive the progression of vascular diseases including diabetic retinopathy. Therefore, the EG presents a novel therapeutic target for treatment of vascular complications. In this review article, we provide an overview of the structure and function of the EG in the retina. Our particular focus is on hyperglycemia-induced perturbations in the glycocalyx structure in the retina, potential underlying mechanisms, and clinical trials studying protective treatments against degradation of the EG.
Collapse
Affiliation(s)
- Gaganpreet Kaur
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States
| | - Norman R Harris
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States
| |
Collapse
|
8
|
Mohammad DN, Ibraheem BF, Khudair HH, Mahmood DK. Expression of Syndecan-1 and Cyclin D1 in Salivary Gland Tumors in Relation to Clinicopathological Parameters. Int J Gen Med 2023; 16:823-835. [PMID: 36883123 PMCID: PMC9985872 DOI: 10.2147/ijgm.s401747] [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: 12/29/2022] [Accepted: 02/10/2023] [Indexed: 03/05/2023] Open
Abstract
Background Salivary tumors have various morphological features and might share some histopathological findings. They are considered a problematic area in diagnosis due to complex clinicopathological features and different biological behavior. Objective To identify the pathological behavior of salivary tumors immunohistochemically. Methodology This retrospective study involved thirty formalin-fixed paraffin-embedded blocks of salivary gland tumors. These tumors were stained immunohistochemically with syndecan-1 and cyclin D1. Chi-Square test was used to relate immunoscoring, intracellular localization, intensity, and invasion to different salivary tumors. The correlation of these two markers was done by spearman's rho test. P-value <0.05 was considered statistically significant. Results The mean age of the patients was 48.69 ± 17.7. The parotid gland was the most commonly reported site in benign tumors, and regarding malignant tumors, maxilla was the most prevalent site. Syndecan-1 in benign tumors showed a predominate score 3, most widely detected in pleomorphic adenoma. Malignant salivary tumors showed 89.4% positive expression with a more frequent score 3, most commonly found in adenocystic carcinoma. Cyclin D1 expressed in all benign salivary tumors, with prominent diffuse mixed intracellular localization in pleomorphic adenoma. Malignant tumors revealed an expression of 94.7%. Moderate scoring with mixed intracellular localization was recorded in adenocystic carcinoma, followed by mucoepidermoid carcinoma. There was a significant correlation between the two markers in response to the distribution of immunostaining in different cell compartments. Conclusion Syndecan-1 and cyclin D1 showed a significant combined role in salivary tumor progression. Interestingly notable ductal-myoepithelial cells affect epithelial morphogenesis, and growth of pleomorphic adenoma was observed. Furthermore, basophilic cells of cribriform adenocystic carcinomas might control the aggressiveness and proliferation rate of these tumors.
Collapse
Affiliation(s)
- Dena Nadhim Mohammad
- Oral Pathology, Oral Diagnosis Department, College of Dentistry, University of Sulaimani, Sulaimani, Iraq
| | - Ban Falih Ibraheem
- Oral Pathology, Oral Diagnosis Department, College of Dentistry, University of Sulaimani, Sulaimani, Iraq
| | - Hassanain Hafidh Khudair
- General Pathology, Pathology Department, College of Medicine, University of Sulaimani, Sulaimani, Iraq
| | - Darya Khalid Mahmood
- Oral Pathology, Oral Diagnosis Department, College of Dentistry, University of Sulaimani, Sulaimani, Iraq
| |
Collapse
|
9
|
Gui XY, Rabkin SW. C-Reactive Protein, Interleukin-6, Trimethylamine-N-Oxide, Syndecan-1, Nitric Oxide, and Tumor Necrosis Factor Receptor-1 in Heart Failure with Preserved Versus Reduced Ejection Fraction: a Meta-Analysis. Curr Heart Fail Rep 2023; 20:1-11. [PMID: 36479675 DOI: 10.1007/s11897-022-00584-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/05/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review was to synthesize the evidence on non-traditional biomarkers from proteomic and metabolomic studies that may distinguish heart failure (HF) with preserved ejection fraction (HFpEF) from heart failure with reduced ejection fraction (HFrEF) and non-HF. RECENT FINDINGS Understanding the pathophysiology of HFpEF continues to be challenging. A number of inflammatory and metabolic biomarkers that have recently been suggested to be involved include C-reactive protein (CRP), interleukin-6 (IL-6), trimethylamine-N-oxide (TMAO), syndecan-1 (SDC-1), nitric oxide (NO), and tumor necrosis factor receptor-1 (TNFR-1). A systematic search was conducted using Medline, EMBASE, and Web of Science with search terms such as "HFpEF," "metabolomics," and "proteomics," and a meta-analysis was conducted. The results demonstrate significantly higher levels of TMAO, CRP, SDC-1, and IL-6 in HFpEF compared to controls without HF and significantly higher levels of TMAO and CRP in HFrEF compared to controls. The results further suggest that HFpEF might be distinguishable from HFrEF based on higher levels of IL-6 and lower levels of SDC-1 and NO. These data may reflect pathophysiological differences between HFpEF and HFrEF.
Collapse
Affiliation(s)
- Xi Yao Gui
- Department of Medicine, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Simon W Rabkin
- Department of Medicine, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.
- Division of Cardiology, University of British Columbia, 9Th Floor 2775 Laurel St, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|
10
|
The Role of Membrane-Type 1 Matrix Metalloproteinase-Substrate Interactions in Pathogenesis. Int J Mol Sci 2023; 24:ijms24032183. [PMID: 36768503 PMCID: PMC9917210 DOI: 10.3390/ijms24032183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023] Open
Abstract
A protease is an enzyme with a proteolytic activity that facilitates the digestion of its substrates. Membrane-type I matrix metalloproteinase (MT1-MMP), a member of the broader matrix metalloproteinases (MMP) family, is involved in the regulation of diverse cellular activities. MT1-MMP is a very well-known enzyme as an activator of pro-MMP-2 and two collagenases, MMP-8 and MMP-13, all of which are essential for cell migration. As an anchored membrane enzyme, MT1-MMP has the ability to interact with a diverse group of molecules, including proteins that are not part of the extracellular matrix (ECM). Therefore, MT1-MMP can regulate various cellular activities not only by changing the extra-cellular environment but also by regulating cell signaling. The presence of both intracellular and extra-cellular portions of MT1-MMP can allow it to interact with proteins on both sides of the cell membrane. Here, we reviewed the MT1-MMP substrates involved in disease pathogenesis.
Collapse
|
11
|
Heparan Sulfate Glycosaminoglycan Is Predicted to Stabilize Inflammatory Infiltrate Formation and RANKL/OPG Ratio in Severe Periodontitis in Humans. Bioengineering (Basel) 2022; 9:bioengineering9100566. [DOI: 10.3390/bioengineering9100566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Since chronically inflamed periodontal tissue exhibits extracellular matrix (ECM) degradation, the possible alternative to standard periodontitis treatment is to restore ECM by supplementing its components, including heparan sulfate glycosaminoglycan (HS GAG). Supplementation of the degraded ECM with synthetic derivatives of HS GAGs has been shown to be effective for periodontal tissue regeneration in experimental animal models of periodontitis. However, the potential of HS GAG supplementation for the treatment of periodontal disease in humans is still unknown. Here, we used a statistical model to investigate the role of HS GAG on inflammatory infiltrate formation and alveolar bone resorption in humans with severe periodontitis. The model was based on data from immunofluorescence staining (IF) of human gingiva samples, and reconstruction of a subset of HS GAG -related proteins from STRING reactome database. According to predictions, increased expression of native HS GAG might stabilize the accumulation of gingival inflammatory infiltrate (represented by the general inflammatory cell marker CD45) and alveolar bone resorption (represented by Receptor Activator of Nuclear ΚΒ ligand (RANKL) and osteoprotegerin (OPG) ratio) but could not restore them to healthy tissue levels. Therefore, supplementation of native HS GAG may be of limited benefits for the treatment of sever periodontitis in humans.
Collapse
|
12
|
Cessna H, Baritaki S, Zaravinos A, Bonavida B. The Role of RKIP in the Regulation of EMT in the Tumor Microenvironment. Cancers (Basel) 2022; 14:cancers14194596. [PMID: 36230521 PMCID: PMC9559516 DOI: 10.3390/cancers14194596] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Raf kinase inhibitor protein (RKIP) expression in cancer cells is significantly reduced and promoting cancer cells growth and invasiveness. Overexpresssion of RKIP has been reported to mediate pleiotropic anti-cancer activities including the inhibition of survival signaling pathways, sensitization to cell death by cytotoxic drugs, inhibition of invasion, EMT and metastasis. The molecular mechanism by which RKIP inhibits EMT is not clear. In this review, we have examined how RKIP inhibits the selected EMT gene products (Snail, vimentin, N-cadherin, laminin alpha) and found that it involves signaling cross-talks between RKIP and each of the EMT gene products. These findings were validated by bioinformatic analyses demonstrating in various human cancers a negative correlation between the expression of RKIP and the expression of the EMT gene products. These findings suggest that targeting RKIP induction in cancer cells will result in multiple hits by inhibiting tumor growth, metastasis and reversal of chemo-immuno resistance. Abstract The Raf Kinase Inhibitor Protein (RKIP) is a unique gene product that directly inhibits the Raf/Mek/Erk and NF-kB pathways in cancer cells and resulting in the inhibition of cell proliferation, viability, EMT, and metastasis. Additionally, RKIP is involved in the regulation of cancer cell resistance to both chemotherapy and immunotherapy. The low expression of RKIP expression in many cancer types is responsible, in part, for the pathogenesis of cancer and its multiple properties. The inhibition of EMT and metastasis by RKIP led to its classification as a tumor suppressor. However, the mechanism by which RKIP mediates its inhibitory effects on EMT and metastases was not clear. We have proposed that one mechanism involves the negative regulation by RKIP of the expression of various gene products that mediate the mesenchymal phenotype as well as the positive regulation of gene products that mediate the epithelial phenotype via signaling cross talks between RKIP and each gene product. We examined several EMT mesenchymal gene products such as Snail, vimentin, N-cadherin, laminin and EPCAM and epithelial gene products such as E-cadherin and laminin. We have found that indeed these negative and positive correlations were detected in the signaling cross-talks. In addition, we have also examined bioinformatic data sets on different human cancers and the findings corroborated, in large part, the findings observed in the signaling cross-talks with few exceptions in some cancer types. The overall findings support the underlying mechanism by which the tumor suppressor RKIP regulates the expression of gene products involved in EMT and metastasis. Hence, the development of agent that can selectively induce RKIP expression in cancers with low expressions should result in the activation of the pleiotropic anti-cancer activities of RKIP and resulting in multiple effects including inhibition of tumor cell proliferation, EMT, metastasis and sensitization of resistant tumor cells to respond to both chemotherapeutics and immunotherapeutics.
Collapse
Affiliation(s)
- Hannah Cessna
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Stavroula Baritaki
- Laboratory of Experimental Oncology, Division of Surgery, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 2404, Cyprus
- Basic and Translational Cancer Research Center (BTCRC), Cancer Genetics, Genomics and Systems Biology Laboratory, Nicosia 1516, Cyprus
| | - Benjamin Bonavida
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90095, USA
- Correspondence:
| |
Collapse
|
13
|
Mayer M, Nachtsheim L, Hoffmann F, von Eggeling F, Guntinas-Lichius O, Prinz J, Klußmann JP, Quaas A, Arolt C, Wolber P. CD138 Is Expressed in Different Entities of Salivary Gland Cancer and Their Lymph Node Metastases and Therefore Represents a Potential Therapeutic Target. Int J Mol Sci 2022; 23:ijms23169037. [PMID: 36012301 PMCID: PMC9409357 DOI: 10.3390/ijms23169037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 12/12/2022] Open
Abstract
Advanced salivary gland carcinomas (SGC) often lack therapeutic options. Agents targeting CD138 have recently shown promising results in clinical trials for multiple myeloma and a preclinical trial for triple-negative breast cancer. Immunohistochemistry for CD138 was performed for all patients who had undergone primary surgery for SGC with curative intent. Findings were validated using matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) imaging. Overall, 111 primary SGC and 13 lymph node metastases from salivary duct carcinomas (SaDu) were evaluated. CD138 expression was found in 60% of all SGC with differing expression across entities (p < 0.01). A mean of 25.2% of the tumor cells in mucoepidermoid carcinoma (MuEp) were positive, followed by epithelial-myoepithelial carcinoma (20.9%), acinic cell carcinoma (16.0%), and SaDu (15.2%). High-/intermediate-grade MuEp showed CD138 expression in a mean of 34.8% of tumor cells. For SaDu, lymph node metastases showed CD138 expression in a mean of 31.2% of tumor cells which correlated with CD138 expression in their primaries (p = 0.01; Spearman’s ρ = 0.71). MALDI-MS imaging confirmed the presence of the CD138 protein in SGC. No significant association was found between clinicopathological data, including progression-free survival (p = 0.50) and CD138 expression. CD138 is expressed in the cell membrane of different entities of SGC and SaDu lymph node metastases and therefore represents a potential target for CD138 targeting drugs.
Collapse
Affiliation(s)
- Marcel Mayer
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50931 Cologne, Germany
- Correspondence: ; Tel.: +49-22147877840
| | - Lisa Nachtsheim
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Franziska Hoffmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
- Department of Otorhinolaryngology, MALDI Imaging and Innovative Biophotonics, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Ferdinand von Eggeling
- Department of Otorhinolaryngology, Head and Neck Surgery, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
- Department of Otorhinolaryngology, MALDI Imaging and Innovative Biophotonics, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Orlando Guntinas-Lichius
- Department of Otorhinolaryngology, Head and Neck Surgery, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
| | - Johanna Prinz
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, 50937 Cologne, Germany
| | - Jens Peter Klußmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Alexander Quaas
- Medical Faculty, Institute of Pathology, University of Cologne, 50937 Cologne, Germany
| | - Christoph Arolt
- Medical Faculty, Institute of Pathology, University of Cologne, 50937 Cologne, Germany
| | - Philipp Wolber
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
14
|
George K, Poudel P, Chalasani R, Goonathilake MR, Waqar S, George S, Jean-Baptiste W, Yusuf Ali A, Inyang B, Koshy FS, Mohammed L. A Systematic Review of Maternal Serum Syndecan-1 and Preeclampsia. Cureus 2022; 14:e25794. [PMID: 35836437 PMCID: PMC9273188 DOI: 10.7759/cureus.25794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/09/2022] [Indexed: 11/22/2022] Open
Abstract
Exploration of novel biomarkers has been gaining popularity in preeclampsia, which is currently being diagnosed based on clinical criteria alone. Soluble syndecan-1, released from one of the proteoglycans associated with the syncytiotrophoblastic layer of the placenta, is affected in patients with abnormal placentation. This article is the first systematic literature review that evaluates the relationship between the antepartum serum levels of the syndecan-1 and preeclampsia. Eight studies were selected after screening and quality appraisal, and data were analyzed. The serum concentration of syndecan-1 was found to correlate positively with the gestational age in all pregnancies and negatively with the systolic blood pressure in patients with preeclampsia. Extremely low levels of soluble syndecan-1 may be helpful as a predictor for the development of preeclampsia during gestation.
Collapse
Affiliation(s)
- Kitty George
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Prakar Poudel
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Roopa Chalasani
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | | | - Sara Waqar
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sheeba George
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Wilford Jean-Baptiste
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Amina Yusuf Ali
- Pediatrics, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Bithaiah Inyang
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Feeba Sam Koshy
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Lubna Mohammed
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
15
|
Ayoub NM, Jaradat SK, Al-Shami KM, Alkhalifa AE. Targeting Angiogenesis in Breast Cancer: Current Evidence and Future Perspectives of Novel Anti-Angiogenic Approaches. Front Pharmacol 2022; 13:838133. [PMID: 35281942 PMCID: PMC8913593 DOI: 10.3389/fphar.2022.838133] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a vital process for the growth and dissemination of solid cancers. Numerous molecular pathways are known to drive angiogenic switch in cancer cells promoting the growth of new blood vessels and increased incidence of distant metastasis. Several angiogenesis inhibitors are clinically available for the treatment of different types of advanced solid cancers. These inhibitors mostly belong to monoclonal antibodies or small-molecule tyrosine kinase inhibitors targeting the classical vascular endothelial growth factor (VEGF) and its receptors. Nevertheless, breast cancer is one example of solid tumors that had constantly failed to respond to angiogenesis inhibitors in terms of improved survival outcomes of patients. Accordingly, it is of paramount importance to assess the molecular mechanisms driving angiogenic signaling in breast cancer to explore suitable drug targets that can be further investigated in preclinical and clinical settings. This review summarizes the current evidence for the effect of clinically available anti-angiogenic drugs in breast cancer treatment. Further, major mechanisms associated with intrinsic or acquired resistance to anti-VEGF therapy are discussed. The review also describes evidence from preclinical and clinical studies on targeting novel non-VEGF angiogenic pathways in breast cancer and several approaches to the normalization of tumor vasculature by targeting pericytes, utilization of microRNAs and extracellular tumor-associate vesicles, using immunotherapeutic drugs, and nanotechnology.
Collapse
Affiliation(s)
- Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
- *Correspondence: Nehad M. Ayoub,
| | - Sara K. Jaradat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Kamal M. Al-Shami
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Amer E. Alkhalifa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| |
Collapse
|
16
|
Abstract
Macropinocytosis is a critical route of nutrient acquisition in pancreatic cancer cells. Constitutive macropinocytosis is promoted by mutant KRAS, which activates the PI3Kα lipid kinase and RAC1, to drive membrane ruffling, macropinosome uptake and processing. However, our recent study on the KRASG12R mutant indicated the presence of a KRAS-independent mode of macropinocytosis in pancreatic cancer cell lines, thereby increasing the complexity of this process. We found that KRASG12R-mutant cell lines promote macropinocytosis independent of KRAS activity using PI3Kγ and RAC1, highlighting the convergence of regulation on RAC signaling. While macropinocytosis has been proposed to be a therapeutic target for the treatment of pancreatic cancer, our studies have underscored how little we understand about the activation and regulation of this metabolic process. Therefore, this review seeks to highlight the differences in macropinocytosis regulation in the two cellular subtypes while also highlighting the features that make the KRASG12R mutant atypical.
Collapse
Affiliation(s)
- G Aaron Hobbs
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA.
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.
| | - Channing J Der
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
17
|
Cho SF, Xing L, Anderson KC, Tai YT. Promising Antigens for the New Frontier of Targeted Immunotherapy in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13236136. [PMID: 34885245 PMCID: PMC8657018 DOI: 10.3390/cancers13236136] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Defining the specificity and biological sequalae induced by receptors differentiated expressed in multiple myeloma cells are critical for the development of effective immunotherapies based on monoclonal antibodies. Ongoing studies continue to discover new antigens with superior tumor selectivity and defined function in regulating the pathophysiology of myeloma cells directly or indirectly in the immunosuppressive bone marrow microenvironment. Meanwhile, it is urgent to identify mechanisms of immune resistance and design more potent immunotherapies, alone and/or with best combination partners to further prolong anti-MM immunity. Abstract The incorporation of novel agents in recent treatments in multiple myeloma (MM) has improved the clinical outcome of patients. Specifically, the approval of monoclonal antibody (MoAb) against CD38 (daratumumab) and SLAMF7 (elotuzumab) in relapsed and refractory MM (RRMM) represents an important milestone in the development of targeted immunotherapy in MM. These MoAb-based agents significantly induce cytotoxicity of MM cells via multiple effector-dependent mechanisms and can further induce immunomodulation to repair a dysfunctional tumor immune microenvironment. Recently, targeting B cell maturation antigen (BCMA), an even MM-specific antigen, has shown high therapeutic activities by chimeric antigen receptor T cells (CAR T), antibody-drug conjugate (ADC), bispecific T-cell engager (BiTE), as well as bispecific antibody (BiAb), with some already approved for heavily pretreated RRMM patients. New antigens, such as orphan G protein-coupled receptor class C group 5 member D (GPRC5D) and FcRH5, were identified and rapidly moved to ongoing clinical studies. We here summarized the pathobiological function of key MM antigens and the status of the corresponding immunotherapies. The potential challenges and emerging treatment strategies are also discussed.
Collapse
Affiliation(s)
- Shih-Feng Cho
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Lijie Xing
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China;
| | - Kenneth C. Anderson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
- Correspondence: ; Tel.: +1-617-632-3875; Fax: +1-617-632-2140
| |
Collapse
|
18
|
Kaur G, Rogers J, Rashdan NA, Cruz-Topete D, Pattillo CB, Hartson SD, Harris NR. Hyperglycemia-induced effects on glycocalyx components in the retina. Exp Eye Res 2021; 213:108846. [PMID: 34801534 PMCID: PMC8665121 DOI: 10.1016/j.exer.2021.108846] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 11/19/2022]
Abstract
PURPOSE Diabetic retinopathy is a vision-threatening complication of diabetes characterized by endothelial injury and vascular dysfunction. The loss of the endothelial glycocalyx, a dynamic layer lining all endothelial cells, contributes to several microvascular pathologies, including an increase in vascular permeability, leukocyte plugging, and capillary occlusion, and may drive the progression of retinopathy. Previously, a significant decrease in glycocalyx thickness has been observed in diabetic retinas. However, the effects of diabetes on specific components of the retinal glycocalyx have not yet been studied. Therefore, the aim of our study was to investigate changes in synthesis, expression, and shedding of retinal glycocalyx components induced by hyperglycemia, which could provide a novel therapeutic target for diabetic retinopathy. METHODS Primary rat retinal microvascular endothelial cells (RRMECs) were grown under normal glucose (5 mM) or high-glucose (25 mM) conditions for 6 days. The mRNA and protein levels of the glycocalyx components were examined using qRT-PCR and Western blot analysis, respectively. Further, mass spectrometry was used to analyze protein intensities of core proteins. In addition, the streptozotocin-induced Type 1 diabetic rat model was used to study changes in the expression of the retinal glycocalyx in vivo. The shedding of the glycocalyx was studied in both culture medium and in plasma using Western blot analysis. RESULTS A significant increase in the shedding of syndecan-1 and CD44 was observed both in vitro and in vivo under high-glucose conditions. The mRNA levels of syndecan-3 were significantly lower in the RRMECs grown under high glucose conditions, whereas those of syndecan-1, syndecan-2, syndecan-4, glypican-1, glypican-3, and CD44 were significantly higher. The protein expression of syndecan-3 and glypican-1 in RRMECs was reduced considerably following exposure to high glucose, whereas that of syndecan-1 and CD44 increased significantly. In addition, mass spectrometry data also suggests a significant increase in syndecan-4 and a significant decrease in glypican-3 protein levels with high glucose stimulation. In vivo, our data also suggest a significant decrease in the mRNA transcripts of syndecan-3 and an increase in mRNA levels of glypican-1 and CD44 in the retinas of diabetic rats. The diabetic rats exhibited a significant reduction in the retinal expression of syndecan-3 and CD44. However, the expression of syndecan-1 and glypican-1 increased significantly in the diabetic retina. CONCLUSIONS One of the main findings of our study was the considerable diversity of glucose-induced changes in expression and shedding of various components of endothelial glycocalyx, for example, increased endothelial and retinal syndecan-1, but decreased endothelial and retinal syndecan-3. This indicates that the reported decrease in the retinal glycocalyx in diabetes in not a result of a non-specific shedding mechanism. Moreover, mRNA measurements indicated a similar diversity, with increases in endothelial and/or retinal levels of syndecan-1, glypican-1, and CD44, but a decrease for syndecan-3, with these increases in mRNA potentially a compensatory reaction to the overall loss of glycocalyx.
Collapse
Affiliation(s)
- Gaganpreet Kaur
- Louisiana State University Health Science Center-Shreveport, LA, Department of Molecular and Cellular Physiology, USA
| | - Janet Rogers
- Oklahoma State University, OK, Department of Biochemistry and Molecular Biology, USA
| | - Nabil A Rashdan
- Louisiana State University Health Science Center-Shreveport, LA, Department of Molecular and Cellular Physiology, USA
| | - Diana Cruz-Topete
- Louisiana State University Health Science Center-Shreveport, LA, Department of Molecular and Cellular Physiology, USA
| | - Christopher B Pattillo
- Louisiana State University Health Science Center-Shreveport, LA, Department of Molecular and Cellular Physiology, USA
| | - Steven D Hartson
- Oklahoma State University, OK, Department of Biochemistry and Molecular Biology, USA
| | - Norman R Harris
- Louisiana State University Health Science Center-Shreveport, LA, Department of Molecular and Cellular Physiology, USA.
| |
Collapse
|
19
|
Ravi Kiran AVVV, Kusuma Kumari G, Krishnamurthy PT, Khaydarov RR. Tumor microenvironment and nanotherapeutics: intruding the tumor fort. Biomater Sci 2021; 9:7667-7704. [PMID: 34673853 DOI: 10.1039/d1bm01127h] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over recent years, advancements in nanomedicine have allowed new approaches to diagnose and treat tumors. Nano drug delivery systems exploit the enhanced permeability and retention (EPR) effect and enter the tumor tissue's interstitial space. However, tumor barriers play a crucial role, and cause inefficient EPR or the homing effect. Mounting evidence supports the hypothesis that the components of the tumor microenvironment, such as the extracellular matrix, and cellular and physiological components collectively or cooperatively hinder entry and distribution of drugs, and therefore, limit the theragnostic applications of cancer nanomedicine. This abnormal tumor microenvironment plays a pivotal role in cancer nanomedicine and was recently recognized as a promising target for improving nano-drug delivery and their therapeutic outcomes. Strategies like passive or active targeting, stimuli-triggered nanocarriers, and the modulation of immune components have shown promising results in achieving anticancer efficacy. The present review focuses on the tumor microenvironment and nanoparticle-based strategies (polymeric, inorganic and organic nanoparticles) for intruding the tumor barrier and improving therapeutic effects.
Collapse
Affiliation(s)
- Ammu V V V Ravi Kiran
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Garikapati Kusuma Kumari
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Praveen T Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research), Ooty, Tamil Nadu, 643001, India
| | - Renat R Khaydarov
- Institute of Nuclear Physics, Uzbekistan Academy of Sciences, Tashkent, 100047, Uzbekistan.
| |
Collapse
|
20
|
Bangalore H, Carter MJ, Parmar K, Austin C, Shankar-Hari M, Hunt BJ, Tibby SM. Degradation of the Endothelial Glycocalyx Contributes to Metabolic Acidosis in Children Following Cardiopulmonary Bypass Surgery. Pediatr Crit Care Med 2021; 22:e571-e581. [PMID: 33950888 DOI: 10.1097/pcc.0000000000002746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Cardiopulmonary bypass surgery is complicated by metabolic acidosis, microvascular dysfunction, and capillary leak. The glycocalyx-a layer of proteins and sugars lining the vascular endothelium-is degraded during cardiopulmonary bypass. We aimed to describe the kinetics of glycocalyx degradation during and following cardiopulmonary bypass. We hypothesized that cleavage of negatively charged fragments of the glycocalyx would directly induce metabolic acidosis through changes in the strong ion gap (defined using Stewart's physicochemical approach to acid-base chemistry). We also investigated whether glycocalyx degradation was associated with failure of endothelial function and cardiovascular dysfunction. DESIGN Single-center prospective cohort study. SETTING Twenty-two bed surgical/medical PICU. PATIENTS Twenty-seven term infants and children requiring cardiopulmonary bypass surgery for the correction/palliation of congenital heart disease. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We recruited 27 patients, 5 days to 57 months old. We prospectively sampled plasma prior to, during, and following cardiopulmonary bypass at predefined time points. We measured plasma concentrations of interleukin-6 (inflammatory marker), heparan sulfate (negatively charged glycocalyx glycosaminoglycan), and syndecan-1 (neutrally charged glycocalyx protein). We defined the following outcome measures: metabolic acidosis (strong ion gap), renal dysfunction (fold change in creatinine), capillary leak (fluid bolus volume), cardiovascular dysfunction (Vasoactive Inotropic Score), and length of ventilation. In linear regression models, maximum measured heparan sulfate concentration (negatively charged) was associated with metabolic acidosis (p = 0.016), renal dysfunction (p = 0.009), and length of ventilation (p = 0.047). In contrast, maximum measured syndecan-1 concentration (neutrally charged) was not associated with these clinical endpoints (p > 0.30 for all). CONCLUSIONS Our data show that metabolic acidosis (increased strong ion gap) is associated with plasma concentration of heparan sulfate, a negatively charged glycosaminoglycan cleaved from the endothelial glycocalyx during cardiopulmonary bypass. In addition, cleavage of heparan sulfate was associated with renal dysfunction, capillary leak, and global markers of cardiovascular dysfunction. These data highlight the importance of designing translational therapies to protect the glycocalyx in cardiopulmonary bypass.
Collapse
Affiliation(s)
- Harish Bangalore
- Paediatric Intensive Care, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Michael J Carter
- Paediatric Intensive Care, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, St Thomas' Hospital, London, United Kingdom
| | - Kiran Parmar
- Thrombosis and Vascular Biology Research Group, St Thomas' Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Conal Austin
- Department of Cardiology, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
- Institute of Women and Children's Health, King's College London, St Thomas' Hospital, London, United Kingdom
| | - Manu Shankar-Hari
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
- Department of Intensive Care, St Thomas' Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Beverley J Hunt
- Thrombosis and Vascular Biology Research Group, St Thomas' Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Shane M Tibby
- Paediatric Intensive Care, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
- Department of Women and Children's Health, School of Life Course Sciences, King's College London, St Thomas' Hospital, London, United Kingdom
| |
Collapse
|
21
|
Keller-Pinter A, Gyulai-Nagy S, Becsky D, Dux L, Rovo L. Syndecan-4 in Tumor Cell Motility. Cancers (Basel) 2021; 13:cancers13133322. [PMID: 34282767 PMCID: PMC8268284 DOI: 10.3390/cancers13133322] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Cell migration is crucial fReaor metastasis formation and a hallmark of malignancy. The primary cause of high mortality among oncology patients is the ability of cancer cells to metastasize. To form metastasis, primary tumor cells must be intrinsically able to move. The transmembrane, heparan sulfate proteoglycan syndecan-4 (SDC4) exhibits multiple functions in signal transduction by regulating Rac1 GTPase activity and consequently actin remodeling, as well as regulating focal adhesion kinase, protein kinase C-alpha and the level of intracellular calcium. By affecting several signaling pathways and biological processes, SDC4 is involved in cell migration under physiological and pathological conditions as well. In this review, we discuss the SDC4-mediated cell migration focusing on the role of SDC4 in tumor cell movement. Abstract Syndecan-4 (SDC4) is a ubiquitously expressed, transmembrane proteoglycan bearing heparan sulfate chains. SDC4 is involved in numerous inside-out and outside-in signaling processes, such as binding and sequestration of growth factors and extracellular matrix components, regulation of the activity of the small GTPase Rac1, protein kinase C-alpha, the level of intracellular calcium, or the phosphorylation of focal adhesion kinase. The ability of this proteoglycan to link the extracellular matrix and actin cytoskeleton enables SDC4 to contribute to biological functions like cell adhesion and migration, cell proliferation, cytokinesis, cellular polarity, or mechanotransduction. The multiple roles of SDC4 in tumor pathogenesis and progression has already been demonstrated; therefore, the expression and signaling of SDC4 was investigated in several tumor types. SDC4 influences tumor progression by regulating cell proliferation as well as cell migration by affecting cell-matrix adhesion and several signaling pathways. Here, we summarize the general role of SDC4 in cell migration and tumor cell motility.
Collapse
Affiliation(s)
- Aniko Keller-Pinter
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
- Correspondence:
| | - Szuzina Gyulai-Nagy
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
| | - Daniel Becsky
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
| | - Laszlo Dux
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
| | - Laszlo Rovo
- Department of Oto-Rhino-Laryngology and Head-Neck Surgery, University of Szeged, H-6725 Szeged, Hungary;
| |
Collapse
|
22
|
Wu F, Chipman A, Dong JF, Kozar RA. Fibrinogen Activates PAK1/Cofilin Signaling Pathway to Protect Endothelial Barrier Integrity. Shock 2021; 55:660-665. [PMID: 32433215 PMCID: PMC8211399 DOI: 10.1097/shk.0000000000001564] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION We recently demonstrated that fibrinogen stabilizes syndecan-1 on the endothelial cell (EC) surface and contributes to EC barrier protection, though the intracellular signaling pathway remains unclear. P21 (Rac1) activated kinase 1 (PAK1) is a protein kinase involved in intracellular signaling leading to actin cytoskeleton rearrangement and plays an important role in maintaining endothelial barrier integrity. We therefore hypothesized that fibrinogen binding to syndecan-1 activated the PAK1 pathway. METHODS Primary human lung microvascular endothelial cells were incubated in 10% lactated Ringers (LR) solution or 10% fibrinogen saline solution (5 mg/mL). Protein phosphorylation was determined by Western blot analysis and endothelial permeability measured by fluorescein isothiocyanate (FITC)-dextran. Cells were silenced by siRNA transfection. Protein concentration was measured in the lung lavages of mice. RESULTS Fibrinogen treatment resulted in increased syndecan-1, PAK1 activation (phosphorylation), cofilin activation (dephosphorylation), as well as decreased stress fibers and permeability when compared with LR treatment. Cofilin is an actin-binding protein that depolymerizes F-actin to decrease stress fiber formation. Notably, fibrinogen did not influence myosin light chain activation (phosphorylation), a mediator of EC tension. Silencing of PAK1 prevented fibrinogen-induced dephosphorylation of cofilin and barrier integrity. Moreover, to confirm the in vitro findings, mice underwent hemorrhagic shock and were resuscitated with either LR or fibrinogen. Hemorrhage shock decreased lung p-PAK1 levels and caused significant lung vascular leakage. However, fibrinogen administration increased p-PAK1 expression to near sham levels and remarkably prevented the lung leakage. CONCLUSION We have identified a novel pathway by which fibrinogen activates PAK1 signaling to stimulate/dephosphorylate cofilin, leading to disassembly of stress fibers and reduction of endothelial permeability.
Collapse
Affiliation(s)
- Feng Wu
- Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD
| | - Amanda Chipman
- Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD
| | - Jing-Fei Dong
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Rosemary Ann Kozar
- Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
23
|
Syndecans in cancer: A review of function, expression, prognostic value, and therapeutic significance. Cancer Treat Res Commun 2021; 27:100312. [PMID: 33485180 DOI: 10.1016/j.ctarc.2021.100312] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022]
Abstract
While our understanding of tumors and how to treat them has advanced significantly since the days of Aminopterin and the radical mastectomy, cancer remains among the leading causes of death worldwide. Despite innumerable advancements in medical technology the non-static and highly heterogeneous nature of a tumor can make characterization and treatment exceedingly difficult. Because of this complexity, the identification of new cellular constituents that can be used for diagnostic, prognostic, and therapeutic purposes is crucial in improving patient outcomes worldwide. Growing evidence has demonstrated that among the myriad of changes seen in cancer cells, the Syndecan family of proteins has been observed to undergo drastic alterations in expression. Syndecans are transmembrane heparan sulfate proteoglycans that are responsible for cell signaling, proliferation, and adhesion, and many studies have shed light on their unique involvement in both tumor progression and suppression. This review seeks to discuss Syndecan expression levels in various cancers, whether they make reliable biomarkers for detection and prognosis, and whether they may be viable targets for future cancer therapies. The conclusions drawn from the literature reviewed in this article indicate that changes in expression of Syndecan protein can have profound effects on tumor size, metastatic capability, and overall patient survival rate. Further, while data regarding the therapeutic targeting of Syndecan proteins is sparse, the available literature does demonstrate promise for their use in cancer treatment going forward.
Collapse
|
24
|
Olsavszky V, Sticht C, Schmid CD, Winkler M, Wohlfeil SA, Olsavszky A, Schledzewski K, Géraud C, Goerdt S, Leibing T, Koch PS. Exploring the transcriptomic network of multi-ligand scavenger receptor Stabilin-1- and Stabilin-2-deficient liver sinusoidal endothelial cells. Gene 2020; 768:145284. [PMID: 33130055 DOI: 10.1016/j.gene.2020.145284] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/09/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
The Class H scavenger receptors Stabilin-1 (Stab1) and Stabilin-2 (Stab2) are two of the most highly expressed genes in liver sinusoidal endothelial cells (LSECs). While Stab1-deficient (Stab1KO) and Stab2-deficient (Stab2KO) mice are phenotypically unremarkable, Stab1/2-double-deficient (StabDKO) mice exhibit perisinusoidal liver fibrosis, glomerulofibrotic nephropathy and a reduced life expectancy. These conditions are caused by insufficiently scavenged circulating noxious blood factors. The effects of either Stab-single- or double-deficiency on LSEC differentiation and function, however, have not yet been thoroughly investigated. Therefore, we performed comprehensive transcriptomic analyses of primary LSECs from Stab1KO, Stab2KO and StabDKO mice. Microarray analysis revealed dysregulation of pathways and genes involved in established LSEC functions while sinusoidal endothelial marker gene expression was grossly unchanged. 82 genes were significantly altered in Stab1KO, 96 genes in Stab2KO and 238 genes in StabDKO compared with controls; 42 genes were found to be commonly dysregulated in all three groups and all of these genes were downregulated. These commonly downregulated genes (CDGs) were categorized as "potential scavengers," "cell adhesion molecules," "TGF-β/BMP-signaling" or "collagen and extracellular matrix (ECM) components". Among CDGs, Colec10, Lumican and Decorin, were the most strongly down-regulated genes and the corresponding proteins impact on the interaction of LSECs with chemokines, ECM components and carbohydrate structures. Similarly, "chemokine signaling," "cytokine-cytokine receptor interaction" and "ECM-receptor interaction," were the GSEA categories which represented most of the downregulated genes in Stab1KO and Stab2KO LSECs. In summary, our data show that loss of a single Stabilin scavenger receptor - and to a greater extent of both receptors - profoundly alters the transcriptomic repertoire of LSECs. These alterations may affect LSEC-specific functions, especially interactions of LSECs with the ECM and during inflammation as well as clearance of the peripheral blood.
Collapse
Affiliation(s)
- Victor Olsavszky
- Department of Dermatology, Venereology and Allergy, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Mannheim 68167, Germany.
| | - Carsten Sticht
- Center for Medical Research, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Christian D Schmid
- Department of Dermatology, Venereology and Allergy, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Mannheim 68167, Germany
| | - Manuel Winkler
- Department of Dermatology, Venereology and Allergy, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Mannheim 68167, Germany
| | - Sebastian A Wohlfeil
- Department of Dermatology, Venereology and Allergy, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Mannheim 68167, Germany; Section of Clinical and Molecular Dermatology, Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ana Olsavszky
- Department of Dermatology, Venereology and Allergy, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Mannheim 68167, Germany; Section of Clinical and Molecular Dermatology, Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kai Schledzewski
- Department of Dermatology, Venereology and Allergy, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Mannheim 68167, Germany
| | - Cyrill Géraud
- Department of Dermatology, Venereology and Allergy, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Mannheim 68167, Germany; Section of Clinical and Molecular Dermatology, Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Sergij Goerdt
- Department of Dermatology, Venereology and Allergy, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Mannheim 68167, Germany; European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Thomas Leibing
- Department of Dermatology, Venereology and Allergy, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Mannheim 68167, Germany; Section of Clinical and Molecular Dermatology, Department of Dermatology, Venereology and Allergology, University Medical Center and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philipp-Sebastian Koch
- Department of Dermatology, Venereology and Allergy, University Medical Center and Medical Faculty Mannheim, University of Heidelberg, and Center of Excellence in Dermatology, Mannheim 68167, Germany; European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| |
Collapse
|
25
|
Syndecan-1 Promotes Hepatocyte-Like Differentiation of Hepatoma Cells Targeting Ets-1 and AP-1. Biomolecules 2020; 10:biom10101356. [PMID: 32977498 PMCID: PMC7598270 DOI: 10.3390/biom10101356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 01/10/2023] Open
Abstract
Syndecan-1 is a transmembrane heparan sulfate proteoglycan which is indispensable in the structural and functional integrity of epithelia. Normal hepatocytes display strong cell surface expression of syndecan-1; however, upon malignant transformation, they may lose it from their cell surfaces. In this study, we demonstrate that re-expression of full-length or ectodomain-deleted syndecan-1 in hepatocellular carcinoma cells downregulates phosphorylation of ERK1/2 and p38, with the truncated form exerting an even stronger effect than the full-length protein. Furthermore, overexpression of syndecan-1 in hepatoma cells is associated with a shift of heparan sulfate structure toward a highly sulfated type specific for normal liver. As a result, cell proliferation and proteolytic shedding of syndecan-1 from the cell surface are restrained, which facilitates redifferentiation of hepatoma cells to a more hepatocyte-like phenotype. Our results highlight the importance of syndecan-1 in the formation and maintenance of differentiated epithelial characteristics in hepatocytes partly via the HGF/ERK/Ets-1 signal transduction pathway. Downregulation of Ets-1 expression alone, however, was not sufficient to replicate the phenotype of syndecan-1 overexpressing cells, indicating the need for additional molecular mechanisms. Accordingly, a reporter gene assay revealed the inhibition of Ets-1 as well as AP-1 transcription factor-induced promoter activation, presumably an effect of the heparan sulfate switch.
Collapse
|
26
|
Li T, Yang W, Ramadan S, Huang X. Synthesis of O-Sulfated Human Syndecan-1-like Glyco-polypeptides by Incorporating Peptide Ligation and O-Sulfated Glycopeptide Cassette Strategies. Org Lett 2020; 22:6429-6433. [PMID: 32806172 PMCID: PMC7517924 DOI: 10.1021/acs.orglett.0c02243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A successful synthesis of O-sulfated syndecan-1-like (Q23-E120) glyco-polypeptide was accomplished. The synthesis features the integration of an O-sulfated carbohydrate-bearing glycopeptide cassette with efficient protein ligation strategies, overcoming the acid lability of carbohydrate sulfates as a major hurdle in solid-phase peptide synthesis. Crucial to the synthesis is the microwave-assisted Ag(I) ligation, which afforded the ligation product in improved overall yield. This O-sulfated syndecan-1 (Q23-E120) is the longest O-sulfated glyco-polypeptide prepared to date.
Collapse
Affiliation(s)
- Tianlu Li
- National Glycoengineering Research Center, Shandong University, Qingdao, Shandong 266237, China
- Departments of Chemistry and Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Weizhun Yang
- Departments of Chemistry and Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Sherif Ramadan
- Departments of Chemistry and Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Chemistry Department, Faculty of Science, Benha University, Benha, Qaliobiya 13518, Egypt
| | - Xuefei Huang
- Departments of Chemistry and Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
27
|
Cheng YC, Ku WC, Tseng TT, Wu CP, Li M, Lee SC. Anchorage independence altered vasculogenic phenotype of melanoma cells through downregulation in aminopeptidase N /syndecan-1/integrin β4 axis. Aging (Albany NY) 2020; 12:16803-16819. [PMID: 32756007 PMCID: PMC7521491 DOI: 10.18632/aging.103425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/20/2020] [Indexed: 01/24/2023]
Abstract
The detachment of tumor cells from extracellular matrix and survival under anchorage-independence were recognized as the initial step of tumor metastasis. Previously we had demonstrated that anchorage-independence altered gene expressions and showed characteristics of cell invasiveness loss, enhanced chemosensitivity, and enhanced subcutaneous tumor formation. However, whether it affected histological phenotypes in tumor tissues remained unclear. Melanoma metastases were generated in nude mice using adherent or suspended melanoma cells. Examination of melanoma metastases revealed histological features of extensive vascular structures in adherent cell-derived tumors, while not seen in suspended cell-derived tumors. Quantitative proteomic analysis at adherent, suspended, and re-attached melanoma cells suggested that aminopeptidase N was potentially downregulated upon cell suspension or reattachment. Downregulation of aminopeptidase N by gene-specific shRNAs showed reduced cell invasiveness and enhanced subcutaneous tumor formation that was consistent with previous observations. Experiments by suppression or overexpression of aminopeptidase N expression demonstrated that aminopeptidase N regulated syndecan-1 and integrin β4 expression through PKCδ pathway. Histological analysis at melanoma metastases further suggested that CD31+/aminopeptidase N+/syndecan-1+/integrin β4+ phenotypes were associated with vascular structures. In summary, we suggested the expression axis of aminopeptidase N/syndecan-1/integrin β4 in melanoma cells was suppressed by detachment stress, which diminished vascular phenotypes of melanoma metastases.
Collapse
Affiliation(s)
- Yu-Che Cheng
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan,Proteomics Laboratory, Cathay Medical Research Institute, Cathay General Hospital, Taipei, Taiwan,Department of Biomedical Science and Engineering, National Central University, Jhongli, Taiwan
| | - Wei-Chi Ku
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
| | - Ting-Ting Tseng
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
| | - Ching-Po Wu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
| | - Mengjin Li
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
| | - Shao-Chen Lee
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
| |
Collapse
|
28
|
Heparan sulfate loaded polycaprolactone-hydroxyapatite scaffolds with 3D printing for bone defect repair. Int J Biol Macromol 2020; 148:153-162. [DOI: 10.1016/j.ijbiomac.2020.01.109] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/10/2020] [Accepted: 01/10/2020] [Indexed: 01/15/2023]
|
29
|
Abstract
Pleiotrophin (PTN) is a potent mitogenic cytokine with a high affinity for the polysaccharide glycosaminoglycan (GAG). Although it is most strongly associated with neural development during embryogenesis and the neonatal period, its expression has also been linked to a plethora of other physiological events including cancer metastasis, angiogenesis, bone development, and inflammation. A considerable amount of research has been carried out to understand the mechanisms by which PTN regulates these events. In particular, PTN has now been shown to bind a diverse collection of receptors including many GAG-containing proteoglycans. These interactions lead to the activation of many intracellular kinases and, ultimately, activation and transformation of cells. Structural studies of PTN in complex with both GAG and domains from its non-proteoglycan receptors reveal a binding mechanism that relies on electrostatic interactions and points to PTN-induced receptor oligomerization as one of the possible ways PTN uses to control cellular functions.
Collapse
|
30
|
Listik E, Toma L. Glypican-1 in human glioblastoma: implications in tumorigenesis and chemotherapy. Oncotarget 2020; 11:828-845. [PMID: 32180897 PMCID: PMC7061737 DOI: 10.18632/oncotarget.27492] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/29/2020] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is one of the most common malignant brain tumors, with which patients have a mean survival of 24 months. Glypican-1 has been previously shown to be overexpressed in human glioblastoma and to be negatively correlated with patient’s survival. This study aimed to investigate how glypican-1 influences the tumoral profile of human glioblastoma using in vitro cell line models. By downregulating the expression of glypican-1 in U-251 MG cells, we observed that the cellular growth and proliferation were highly reduced, in which cells were significantly shifted towards G0 as opposed to G1 phases. Cellular migration was severely affected, and glypican-1 majorly impacted the affinity towards laminin-binding of glioblastoma U-251 MG cells. This proteoglycan was highly prevalent in glioblastoma cells, being primarily localized in the cellular membrane and extracellular vesicles, occasionally with glypican-3. Glypican-1 could also be found in cell-cell junctions with syndecan-4 but was not identified in lipid rafts in this study. Glypican-1-silenced cells were much more susceptible to temozolomide than in U-251 MG itself. Therefore, we present evidence not only to support facts that glypican-1 is an elementary macromolecule in glioblastoma tumoral microenvironment but also to introduce this proteoglycan as a promising therapeutic target for this lethal tumor.
Collapse
Affiliation(s)
- Eduardo Listik
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Leny Toma
- Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
31
|
Farfán N, Orellana-Serradell O, Herrera D, Chrzanowsky D, Cubillos P, Marín G, Antonio García De Herreros A, Castellón EA, Contreras HR. SNAIL expression correlates with the translocation of syndecan‑1 intracellular domain into the nucleus in prostate cancer cell lines. Int J Mol Med 2020; 45:1073-1080. [PMID: 32124938 PMCID: PMC7053857 DOI: 10.3892/ijmm.2020.4488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/21/2020] [Indexed: 01/06/2023] Open
Abstract
Zinc finger protein SNAI1 (SNAIL) and zinc finger protein SNAI2 (SLUG) transcription factors promote epithelial-mesenchymal transition, a process through which epithelial cells acquire a mesenchymal phenotype, increasing their migratory and invasive properties. In prostate cancer (PCa) progression, increased expression levels of SNAIL and SLUG have been described. In advanced PCa, a decrease in the cell surface proteoglycan syndecan-1 (SDC-1), which has a role in cell-to-extracellular matrix adhesion, has been observed. Notably, SDC-1 nuclear location has been observed in mesenchymal cancers. The present study aimed to determine if SNAIL and SLUG may be associated with the nuclear location of SDC-1 in PCa. To determine the location of SDC-1, antibodies against its intracellular domain (ID) or extracellular domain (ED) were used in benign prostatic hyperplasia (BPH) and PCa samples with high Gleason scores. Only ID-SDC-1 was located in the cell nuclei in advanced PCa samples, but not in the BPH samples. ED-SDC-1 was located in the cell membrane and cytoplasm, exhibiting decreased levels in PCa in comparison with those in BPH. Furthermore, LNCaP and PC3 PCa cell lines with ectopic SNAIL expression exhibited nuclear ID-SDC-1. No change was observed in the ED-SDC-1 levels, and maintained its location in the cell membrane and cytoplasm. SLUG induced no change in ID-SDC-1 location. At the protein level, an association between SNAIL and nuclear ID-SDC-1 was observed. In conclusion, the results of the present study demonstrated that nuclear ID-SDC-1 localization was associated with SNAIL expression in PCa cell lines.
Collapse
Affiliation(s)
- Nancy Farfán
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Octavio Orellana-Serradell
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Daniela Herrera
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Dominique Chrzanowsky
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Paulina Cubillos
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Gabriel Marín
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | | | - Enrique A Castellón
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Héctor R Contreras
- Department of Basic and Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| |
Collapse
|
32
|
Miftode RS, Şerban IL, Timpau AS, Miftode IL, Ion A, Buburuz AM, Costache AD, Costache II. Syndecan-1: A Review on Its Role in Heart Failure and Chronic Liver Disease Patients' Assessment. Cardiol Res Pract 2019; 2019:4750580. [PMID: 31815014 PMCID: PMC6878788 DOI: 10.1155/2019/4750580] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/18/2019] [Accepted: 08/30/2019] [Indexed: 12/13/2022] Open
Abstract
The close connection and interaction between the cardiac and the liver functions are well-known, as cirrhotic cardiomyopathy is an important clinical entity which best describes the mutual pathogenical influence between these two organs. Due to the fact that cardiac dysfunction in patients with chronic hepatic disorders is oligosymptomatic or even asymptomatic, an early diagnosis represents a challenge for every physician. Syndecan-1-a transmembrane proteoglycan that exerts its functions mainly via its heparane sulfate chains-is a very promising biomarker, correlated not only with the degree of cardiac fibrosis but also with the severity of liver fibrosis. Many studies highlighted its role in the development of cardiac fibrosis or atherogenesis, being significantly correlated with the activity of angiotensin II. Multiple evidence revealed that syndecan-1 is also associated with tissue injury and may regulate inflammatory and regenerative responses, being considered a protective molecule that limits the inflammation and reduces cardiac remodelling and dysfunction after a myocardial infarction. Syndecan-1 may also be used as a reliable biomarker for the noninvasive assessment of liver fibrosis. Under various fibrogenetic conditions, shedding of syndecan's extracellular domain took place, becoming a soluble form that binds different growth factors and inhibits further fibrosis. This complex molecule is also involved in the lipid metabolism, by altering the clearance of cholesterol particles, and in chronic hepatitis, by enhancing the viral invasion of hepatocytes. Due to the growing interest in this biomarker, multiple studies aimed at revealing syndecan-1's potential benefits in the diagnosis and prognosis assessment in patients with heart failure or chronic liver disorders. In this review, we review the mechanisms by which syndecan-1 exerts its effects and the possible perspectives opened by its use as a dual cardio-hepatic biomarker.
Collapse
Affiliation(s)
- Radu-Stefan Miftode
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, Iasi 700115, Romania
| | - Ionela-Lăcrămioara Şerban
- Department of Morpho-Functional Sciences (II), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, Iasi 700115, Romania
| | - Amalia-Stefana Timpau
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, Iasi 700115, Romania
| | - Ionela-Larisa Miftode
- Department of Infectious Diseases, Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, Iasi 700115, Romania
| | - Adriana Ion
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, Iasi 700115, Romania
| | - Ana-Maria Buburuz
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, Iasi 700115, Romania
| | - Alexandru-Dan Costache
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, Iasi 700115, Romania
| | - Irina-Iuliana Costache
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, Iasi 700115, Romania
| |
Collapse
|
33
|
Zandonadi FS, Castañeda Santa Cruz E, Korvala J. New SDC function prediction based on protein-protein interaction using bioinformatics tools. Comput Biol Chem 2019; 83:107087. [PMID: 31351242 DOI: 10.1016/j.compbiolchem.2019.107087] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 05/13/2019] [Accepted: 06/23/2019] [Indexed: 12/11/2022]
Abstract
The precise roles for SDC have been complex to specify. Assigning and reanalyzing protein and peptide identification to novel protein functions is one of the most important challenges in postgenomic era. Here, we provide SDC molecular description to support, contextualize and reanalyze the corresponding protein-protein interaction (PPI). From SDC-1 data mining, we discuss the potential of bioinformatics tools to predict new biological rules of SDC. Using these methods, we have assembled new possibilities for SDC biology from PPI data, once, the understanding of biology complexity cannot be capture from one simple question.
Collapse
Affiliation(s)
- Flávia S Zandonadi
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Departamento de Química Analítica, Universidade de Campinas, UNICAMP, Campinas, SP, Brazil.
| | - Elisa Castañeda Santa Cruz
- Laboratory of Bioanalytics and Integrated Omics (LaBIOmics), Departamento de Química Analítica, Universidade de Campinas, UNICAMP, Campinas, SP, Brazil
| | - Johanna Korvala
- Cancer and Translational Medicine Research Unit, Biocenter Oulu and Faculty of Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
34
|
Glycosylation in cancer: Selected roles in tumour progression, immune modulation and metastasis. Cell Immunol 2018; 333:46-57. [DOI: 10.1016/j.cellimm.2018.03.007] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/13/2018] [Accepted: 03/16/2018] [Indexed: 01/20/2023]
|
35
|
Hempel C, Sporring J, Kurtzhals JAL. Experimental cerebral malaria is associated with profound loss of both glycan and protein components of the endothelial glycocalyx. FASEB J 2018; 33:2058-2071. [DOI: 10.1096/fj.201800657r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Casper Hempel
- Centre for Medical ParasitologyDepartment of Clinical MicrobiologyCopenhagen University HospitalCopenhagenDenmark
- Department of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
- Department of Micro- and NanotechnologyTechnical University of DenmarkLyngbyDenmark
| | - Jon Sporring
- Department for Computer SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Jørgen Anders Lindholm Kurtzhals
- Centre for Medical ParasitologyDepartment of Clinical MicrobiologyCopenhagen University HospitalCopenhagenDenmark
- Department of Immunology and MicrobiologyUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
36
|
Uen Y, Wang JW, Wang C, Jhang Y, Chung JY, Tseng T, Sheu M, Lee S. Mining of potential microRNAs with clinical correlation - regulation of syndecan-1 expression by miR-122-5p altered mobility of breast cancer cells and possible correlation with liver injury. Oncotarget 2018; 9:28165-28175. [PMID: 29963269 PMCID: PMC6021336 DOI: 10.18632/oncotarget.25589] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 05/24/2018] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs are small noncoding RNAs acting as novel biomarkers of various diseases and potential regulators of protein expression and functions. Syndecan-1 is the heparan sulfate proteoglycan associated with malignancy of various cancers, including breast cancer. In this study, we proposed a experimental workflow to investigate potential microRNAs that regulate SDC1 expression and affect breast cancer cell mobility. MicroRNA candidates were selected from available Gene Expression Omnibus datasets on breast malignancy. Further in silico duplex hybridization and multiplex PCR approach were used to screen potential microRNAs. Analysis showed increased syndecan-1 expression but decreased miR-122-5p level upon breast malignancy. Western blot and in vitro luciferase assay confirmed the targeting of 3'-untranslated region of syndecan-1 and suppression of syndecan-1 expression by miR-122-5p. The suppression of syndecan-1 expression by miR-122-5p or shRNAs against syndecan-1 increased breast cancer cell mobility; while overexpression of syndecan-1 inhibited cell mobility. In further, miR-122-5p was enriched in liver cell-derived exosomes that was able to suppress syndecan-1 expression and increase cell mobility in breast cancer cells. In conclusion, our results suggested the downregulation of SDC1 by miR-122-5p or liver-cell-derived exosomes would enhance breast cancer cell mobility. Metastasis or mobility of breast cancer cells might be affected by circulating miR-122-5p and not directly correlated with progression of breast cancer.
Collapse
Affiliation(s)
- YihHuei Uen
- Department of Surgery, ChiMei Hospital, Chiali, Jiali Dist., Tainan City, Taiwan
- Department of Surgery, Asia University Hospital, Taichung City, Taiwan
- Department of Biotechnology, Asia University, Taichung City, Taiwan
| | - Jin-Wun Wang
- Department of Surgery, ChiMei Hospital, Chiali, Jiali Dist., Tainan City, Taiwan
| | - ChiaChen Wang
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City, Taiwan
- Department of Dermatology, Cardinal Tien Hospital, Xindian Dist., New Taipei City, Taiwan
| | - Yaoyun Jhang
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City, Taiwan
| | - Jo-Yun Chung
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City, Taiwan
| | - TingTing Tseng
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City, Taiwan
| | - MingJen Sheu
- Department of Hepato-Gastroenterology, ChiMei Hospital, Yongkang Dist., Tainan City, Taiwan
| | - ShaoChen Lee
- School of Medicine, Fu Jen Catholic University, Xinzhuang Dist., New Taipei City, Taiwan
| |
Collapse
|
37
|
Kero D, Bilandzija TS, Arapovic LL, Vukojevic K, Saraga-Babic M. Syndecans and Enzymes Involved in Heparan Sulfate Biosynthesis and Degradation Are Differentially Expressed During Human Odontogenesis. Front Physiol 2018; 9:732. [PMID: 29962964 PMCID: PMC6010574 DOI: 10.3389/fphys.2018.00732] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/25/2018] [Indexed: 01/01/2023] Open
Abstract
Syndecans belong to a four-member family of cell surface heparan sulfate proteoglycans (HSPGs) abundantly present in various tissues. They are primarily recognized as extracellular matrix (ECM) receptors able to bind various ECM components and form gradients of morphogens and growth factors. Syndecans are composed of core protein with distinctive cytoplasmic, transmembrane, and extracellular domains to which several HS glycosaminoglycan (GAG) chains are covalently attached. In development of composite organs, such as teeth, expression patterns of syndecans display temporo-spatial shifts between epithelial and mesenchymal tissue compartments. Along with diverse functional properties of syndecans and generally large number of their interactors due to HS GAG chain content, this suggests possible involvement of syndecans in modulation of epithelial-to-mesenchymal crosstalk. Functional versatility of syndecans greatly depends upon the biochemical properties of attached HS GAG chains. These are specifically determined during the HS biosynthesis by the combinatorial action of glycosyl-transferases (Exts/EXTs) and bi-functional sulfotransferases (Ndsts/NDSTs), as well as by post-biosynthetic enzymatic cleavage of HS by the only active endoglucuronidase in mammals, heparanase 1 (Hpse1/HPSE1). Matching the essential requirement for HS during organogenesis, null-mutant animals for genes encoding these enzymes display severe developmental anomalies of mineralized tissues (including teeth) with embryonic or perinatal lethality. In this study, we analyzed expression of syndecan HSPGs (syndecans 1, 2, and 4), enzymes involved in HS biosynthesis (EXT1, NDST1, NDST2) and HS cleavage (HPSE1) in human tooth germs during the early stages of odontogenesis. All of the investigated factors displayed temporo-spatial differences in expression patterns, and some of them showed distinctive asymmetries of expression domains. Our findings suggest that these factors might be differentially involved in cellular processes which take place during the early odontogenic sequence in humans.
Collapse
Affiliation(s)
- Darko Kero
- Department of Dental Morphology and Anthropology, Study Program of Dental Medicine, School of Medicine, University of Split, Split, Croatia
| | - Tanja Simic Bilandzija
- Department of Maxillofacial Surgery, University Clinical Hospital Mostar, Mostar, Bosnia and Herzegovina.,Study Program of Dental Medicine, School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina
| | - Lidija Lasic Arapovic
- Study Program of Dental Medicine, School of Medicine, University of Mostar, Mostar, Bosnia and Herzegovina.,Primary Health Care Center Mostar, Mostar, Bosnia and Herzegovina
| | - Katarina Vukojevic
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Split, Croatia
| | - Mirna Saraga-Babic
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, School of Medicine, University of Split, Split, Croatia
| |
Collapse
|
38
|
Szarvas T, Sevcenco S, Módos O, Keresztes D, Nyirády P, Kubik A, Romics M, Kovalszky I, Reis H, Hadaschik B, Shariat SF, Kramer G. Circulating syndecan-1 is associated with chemotherapy-resistance in castration-resistant prostate cancer. Urol Oncol 2018; 36:312.e9-312.e15. [DOI: 10.1016/j.urolonc.2018.03.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 12/29/2017] [Accepted: 03/12/2018] [Indexed: 12/19/2022]
|
39
|
Xue C, Yu DMT, Gherardi S, Koach J, Milazzo G, Gamble L, Liu B, Valli E, Russell AJ, London WB, Liu T, Cheung BB, Marshall GM, Perini G, Haber M, Norris MD. MYCN promotes neuroblastoma malignancy by establishing a regulatory circuit with transcription factor AP4. Oncotarget 2018; 7:54937-54951. [PMID: 27448979 PMCID: PMC5342392 DOI: 10.18632/oncotarget.10709] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/16/2016] [Indexed: 12/11/2022] Open
Abstract
Amplification of the MYCN oncogene, a member of the MYC family of transcriptional regulators, is one of the most powerful prognostic markers identified for poor outcome in neuroblastoma, the most common extracranial solid cancer in childhood. While MYCN has been established as a key driver of malignancy in neuroblastoma, the underlying molecular mechanisms are poorly understood. Transcription factor activating enhancer binding protein-4 (TFAP4) has been reported to be a direct transcriptional target of MYC. We show for the first time that high expression of TFAP4 in primary neuroblastoma patients is associated with poor clinical outcome. siRNA-mediated suppression of TFAP4 in MYCN-expressing neuroblastoma cells led to inhibition of cell proliferation and migration. Chromatin immunoprecipitation assay demonstrated that TFAP4 expression is positively regulated by MYCN. Microarray analysis identified genes regulated by both MYCN and TFAP4 in neuroblastoma cells, including Phosphoribosyl-pyrophosphate synthetase-2 (PRPS2) and Syndecan-1 (SDC1), which are involved in cancer cell proliferation and metastasis. Overall this study suggests a regulatory circuit in which MYCN by elevating TFAP4 expression, cooperates with it to control a specific set of genes involved in tumor progression. These findings highlight the existence of a MYCN-TFAP4 axis in MYCN-driven neuroblastoma as well as identifying potential therapeutic targets for aggressive forms of this disease.
Collapse
Affiliation(s)
- Chengyuan Xue
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Denise M T Yu
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Samuele Gherardi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Jessica Koach
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Giorgio Milazzo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Laura Gamble
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Bing Liu
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Emanuele Valli
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Amanda J Russell
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Wendy B London
- Boston Children's Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Tao Liu
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Belamy B Cheung
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Glenn M Marshall
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia
| | - Giovanni Perini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,CIRI Health Sciences and Technologies University of Bologna, Bologna, Italy
| | - Michelle Haber
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Murray D Norris
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia.,University of New South Wales Centre for Childhood Cancer Research, Sydney, Australia
| |
Collapse
|
40
|
Nikitovic D, Berdiaki A, Spyridaki I, Krasanakis T, Tsatsakis A, Tzanakakis GN. Proteoglycans-Biomarkers and Targets in Cancer Therapy. Front Endocrinol (Lausanne) 2018; 9:69. [PMID: 29559954 PMCID: PMC5845539 DOI: 10.3389/fendo.2018.00069] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/16/2018] [Indexed: 12/18/2022] Open
Abstract
Proteoglycans (PGs), important constituents of the extracellular matrix, have been associated with cancer pathogenesis. Their unique structure consisting of a protein core and glycosaminoglycan chains endowed with fine modifications constitutes these molecules as capable cellular effectors important for homeostasis and contributing to disease progression. Indeed, differential expression of PGs and their interacting proteins has been characterized as specific for disease evolvement in various cancer types. Importantly, PGs to a large extent regulate the bioavailability of hormones, growth factors, and cytokines as well as the activation of their respective receptors which regulate phenotypic diversibility, gene expression and rates of recurrence in specific tumor types. Defining and targeting these effectors on an individual patient basis offers ground for the development of newer therapeutic approaches which may act as either supportive or a substitute treatment to the standard therapy protocols. This review discusses the roles of PGs in cancer progression, developing technologies utilized for the defining of the PG "signature" in disease, and how this may facilitate the generation of tailor-made cancer strategies.
Collapse
Affiliation(s)
- Dragana Nikitovic
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| | - Aikaterini Berdiaki
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| | - Ioanna Spyridaki
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| | - Theodoros Krasanakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, Heraklion, Greece
| | - George N Tzanakakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, Greece
| |
Collapse
|
41
|
Kaminska K, Czarnecka AM, Khan MI, Fendler W, Klemba A, Krasowski P, Bartnik E, Szczylik C. Effects of cell-cell crosstalk on gene expression patterns in a cell model of renal cell carcinoma lung metastasis. Int J Oncol 2017; 52:768-786. [PMID: 29286165 PMCID: PMC5807041 DOI: 10.3892/ijo.2017.4234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 12/04/2017] [Indexed: 01/28/2023] Open
Abstract
The median survival rate of patients with metastatic renal carcinoma is approximately 10 to 12 months, with up to 50% of patients developing metastases in the lung parenchyma. The molecular basis for metastatic development remains unclear. In the present study, we used renal cell carcinoma (RCC) cells and bronchial epithelial cells, representing metastasis target organ cells, conditioned medium and co-culture models to identify specific gene expression changes responsible for cancer cell viability in a metastatic microenvironment. RCC cell proliferation and migration increased when the culture was supplemented with conditioned medium from lung fibroblasts or pleural epithelial cells. Healthy epithelial cells were, in turn, also stimulated with conditioned medium from RCC cell lines. The mitogen-activated protein kinase (MAPK), interleukin (IL)-6, and phosphatidylinositol 4,5-bisphosphate (PIP2) signaling pathways were identified as deregulated upon cell‑cell interaction. Thus, cell-cell communication may contribute to the development of the metastatic niche. The identified deregulated signaling pathways may be considered as potential therapeutic targets in metastatic renal carcinoma.
Collapse
Affiliation(s)
- Katarzyna Kaminska
- Department of Oncology, Military Institute of Medicine, 04‑141 Warsaw, Poland
| | - Anna M Czarnecka
- Department of Oncology, Military Institute of Medicine, 04‑141 Warsaw, Poland
| | - Mohammed Imran Khan
- Department of Oncology, Military Institute of Medicine, 04‑141 Warsaw, Poland
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92‑215 Lodz, Poland
| | - Aleksandra Klemba
- Department of Oncology, Military Institute of Medicine, 04‑141 Warsaw, Poland
| | - Pawel Krasowski
- Department of Oncology, Military Institute of Medicine, 04‑141 Warsaw, Poland
| | - Ewa Bartnik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, 02‑106 Warsaw, Poland
| | - Cezary Szczylik
- Department of Oncology, Military Institute of Medicine, 04‑141 Warsaw, Poland
| |
Collapse
|
42
|
Mytilinaiou M, Nikitovic D, Berdiaki A, Kostouras A, Papoutsidakis A, Tsatsakis AM, Tzanakakis GN. Emerging roles of syndecan 2 in epithelial and mesenchymal cancer progression. IUBMB Life 2017; 69:824-833. [PMID: 28940845 DOI: 10.1002/iub.1678] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/29/2017] [Indexed: 01/04/2023]
Abstract
Syndecan 2 (SDC2) belongs to a four-member family of evolutionary conserved small type I transmembrane proteoglycans consisting of a protein core to which glycosaminoglycan chains are covalently attached. SDC2 is a cell surface heparan sulfate proteoglycan, which is increasingly drawing attention for its distinct characteristics and its participation in numerous cell functions, including those related to carcinogenesis. Increasing evidence suggests that the role of SDC2 in cancer pathogenesis is dependent on cancer tissue origin rendering its use as a biomarker/therapeutic target feasible. This mini review discusses the mechanisms, through which SDC2, in a distinct manner, modulates complex signalling networks to affect cancer progression. © 2017 IUBMB Life, 69(11):824-833, 2017.
Collapse
Affiliation(s)
- Maria Mytilinaiou
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Dragana Nikitovic
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Aikaterini Berdiaki
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Antonis Kostouras
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Antonis Papoutsidakis
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Aristidis M Tsatsakis
- Laboratory of Toxicology, School of Medicine, University of Crete, Heraklion, Greece
| | - George N Tzanakakis
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
43
|
Abstract
Heparin and heparan sulfate glycosaminoglycans are long, linear polysaccharides that are made up of alternating dissacharide sequences of sulfated uronic acid and amino sugars. Unlike heparin, which is only found in mast cells, heparan sulfate is ubiquitously expressed on the cell surface and in the extracellular matrix of all animal cells. These negatively-charged glycans play essential roles in important cellular functions such as cell growth, adhesion, angiogenesis, and blood coagulation. These biomolecules are also involved in pathophysiological conditions such as pathogen infection and human disease. This review discusses past and current methods for targeting these complex biomolecules as a novel therapeutic strategy to treating disorders such as cancer, neurodegenerative diseases, and infection.
Collapse
Affiliation(s)
- Ryan J Weiss
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA 92093-0358, USA
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA 92093-0358, USA
| | - Yitzhak Tor
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0358, USA.
| |
Collapse
|
44
|
Colombe S, Houllier L, Fleurot E, Levallet G, Benhaïm A, Bonnamy PJ, Levallet J. Syndecan 1 represses cell growth and FSH responsiveness in human granulosa cells. Reproduction 2017; 153:797-808. [DOI: 10.1530/rep-17-0074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 03/17/2017] [Accepted: 03/27/2017] [Indexed: 11/08/2022]
Abstract
Albeit devoid of intrinsic catalytic activity, the transmembrane heparan sulphate proteoglycan syndecan 1 plays critical roles in cellular processes such as extracellular matrix crosstalk, cytoskeletal organization, cell spreading, proliferation and differentiation. During the ovarian cycle, the expression of syndecan 1 in granulosa cells shows cyclic variation suggesting that it might fulfil specific roles in follicle development. To investigate its physiological roles on granulosa cells, syndecan 1 was overexpressed in human granulosa cell line KGN which retains features of granulosa cells from small antral follicle such as estradiol (E2) synthesis and low expression of functional FSH receptor (FSHR). We demonstrated that overexpression of syndecan 1 in immature granulosa cells (KGN-SDC1) induces a profound alteration in their intrinsic characteristics including enhanced spreading and attachment, both associated with a reduced growth rate. Flow cytometry analysis revealed that syndecan 1 overexpression increases the percentage of KGN cells in quiescent phase. This partial cell cycle exit is concordant with downregulated levels of CCND1 and CDK4 and upregulated expression of CDK inhibitor CDKN1A. In parallel both unstimulated and FSH-induced E2 synthesis are reduced in KGN-SDC1 through both repression of CYP19A1 and FSHR mRNA associated with decreased levels of potential regulators NR5A1 and ESR2. Additionally, we provide evidence that transient cAMP accumulation reduction in cells overexpressing syndecan 1 is accompanied by an increase in cAMP-hydrolysing PDE activity. Our results demonstrated that syndecan 1 might regulate differentiation of granulosa cells and follicular development by means of various mechanisms involving morphological changes, control of signalling pathways and alterations in gene expressions.
Free French abstract: A French translation of this abstract is freely available at http://www.reproduction-online.org/content/153/6/797/suppl/DC2
Collapse
|
45
|
Szarvas T, Reis H, Vom Dorp F, Tschirdewahn S, Niedworok C, Nyirady P, Schmid KW, Rübben H, Kovalszky I. Soluble syndecan-1 (SDC1) serum level as an independent pre-operative predictor of cancer-specific survival in prostate cancer. Prostate 2016; 76:977-85. [PMID: 27062540 DOI: 10.1002/pros.23186] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/22/2016] [Indexed: 11/08/2022]
Abstract
BACKGROUND PSA-screening detects many cases of clinically non-aggressive prostate cancer (PC) leading to significant overtreatment. Therefore, pre-operatively available prognostic biomarkers are needed to help therapy decisions. Syndecan-1 (SDC1) is a promising prognostic tissue marker in several cancers including PC but serum levels of shedded SDC1-ectodomain (sSDC1) have not been assessed in PC. METHODS A total of 150 patients with PC were included in this study (n = 99 serum samples, n = 103 paraffin-embedded samples (FFPE), n = 52 overlap). SDC1 protein expression and cellular localization was evaluated by immunohistochemistry (IHC), while sSDC1 serum concentrations were measured by ELISA. Serum sSDC1 levels were compared to those of MMP7, which is known to be a protease involved in SDC1 ectodomain-shedding. Clinico-pathological and follow-up data were collected and correlated with SDC1 tissue and serum levels. Disease (PC)-specific (DSS) and overall-survival (OS) were primary endpoints. RESULTS Median follow-up was 167 months in the serum- and 146 months in the FFPE-group. SDC1-reactivity was higher in non-neoplastic prostate glands compared to PC. In addition, cytoplasmatic, but not membranous SDC1 expression was enhanced in PC patients with higher Gleason-score >6 PC (P = 0.016). Soluble SDC1-levels were higher in patients with Gleason-score >6 (P = 0.043) and metastatic disease (P = 0.022) as well as in patients with progressed disease treated with palliative transurethral resection (P = 0.002). In addition, sSDC1 levels were associated with higher MMP7 serum concentration (P = 0.005). In univariable analyses, only sSDC1-levels exhibited a trend to unfavorable DSS (P = 0.077). In a multivariable pre-operative model, high pre-operative sSDC1-level (>123 ng/ml) proved to be an independent marker of adverse OS (P = 0.048) and DSS (P = 0.020). CONCLUSIONS The present study does not confirm the prognostic relevance of SDC1-IHC. The significant higher sSDC1 serum levels in advanced cases of PC, suggest that SDC1 shedding might be involved in PC progression. Additionally, high sSDC1-level proved to be an independent factor of adverse OS and DSS in a multivariable pre-operative model, making evaluation of sSDC1-levels a promising tool for pre-operative risk-stratification and/or therapy monitoring. Prostate 76:977-985, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tibor Szarvas
- Department of Urology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
- Department of Urology, Semmelweis University, Budapest, Hungary
| | - Henning Reis
- Faculty of Medicine, Institute of Pathology, University Duisburg-Essen, Essen, Germany
| | - Frank Vom Dorp
- Department of Urology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | - Stephan Tschirdewahn
- Department of Urology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | - Christian Niedworok
- Department of Urology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | - Peter Nyirady
- Department of Urology, Semmelweis University, Budapest, Hungary
| | - Kurt W Schmid
- Faculty of Medicine, Institute of Pathology, University Duisburg-Essen, Essen, Germany
| | - Herbert Rübben
- Department of Urology, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | - Ilona Kovalszky
- First Institute of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
46
|
Insights into the molecular roles of heparan sulfate proteoglycans (HSPGs—syndecans) in autocrine and paracrine growth factor signaling in the pathogenesis of Hodgkin’s lymphoma. Tumour Biol 2016; 37:11573-11588. [DOI: 10.1007/s13277-016-5118-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 06/09/2016] [Indexed: 12/25/2022] Open
|
47
|
Evaluation of New Biomarkers in the Prediction of Malignant Mesothelioma in Subjects with Environmental Asbestos Exposure. Lung 2016; 194:409-17. [DOI: 10.1007/s00408-016-9868-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 03/21/2016] [Indexed: 10/22/2022]
|
48
|
Mammary serine protease inhibitor and CD138 immunohistochemical expression in ovarian serous and clear cell carcinomas. Tumour Biol 2015; 37:4889-900. [PMID: 26526579 DOI: 10.1007/s13277-015-4333-y] [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: 08/18/2015] [Accepted: 10/26/2015] [Indexed: 02/05/2023] Open
Abstract
This study aims to investigate the immunohistochemical expression of mammary serine protease inhibitor (maspin) and CD138 in primary ovarian high-grade serous carcinomas (HGSC) as compared to low-grade serous carcinomas (LGSC) and clear cell carcinomas and investigate if the studied markers have a correlation to International Federation of Gynaecology and Obstetrics (FIGO) stage, Ki67 proliferation index, and to each other. Maspin cellular location varied significantly between studied groups with only nuclear expression seen in 46.7 % of LGSC group, mixed nuclear and cytoplasmic in 13.3, 28.6, and 20 % of LGSC, HGSC, and clear cell carcinoma, respectively, and was only cytoplasmic in 26.7, 71.4, and 80 % of LGSC, HGSC, and clear cell carcinoma, respectively. Mean maspin and CD138 counts were significantly higher in HGSC and clear cell carcinoma compared to LGSC. Both maspin and CD138 scores varied significantly between studied groups and were positively correlated with adverse prognostic factors in studied carcinomas including FIGO stage and Ki67 proliferation index. Besides, both maspin and CD138 had significant correlation to each other. These findings suggest that epithelial cytoplasmic expression of maspin and CD138 may have a significant role in tumorigenesis in ovarian high-grade serous carcinomas and clear cell carcinomas; these markers may regulate tumor cell proliferation, and their significant correlation to each other may suggest that CD138 probably induces maspin expression to protect tumor growth factors from being lysed by proteolytic enzymes.
Collapse
|
49
|
Fernández-Vega I, García-Suárez O, García B, Crespo A, Astudillo A, Quirós LM. Heparan sulfate proteoglycans undergo differential expression alterations in right sided colorectal cancer, depending on their metastatic character. BMC Cancer 2015; 15:742. [PMID: 26482785 PMCID: PMC4617710 DOI: 10.1186/s12885-015-1724-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022] Open
Abstract
Background Heparan sulfate proteoglycans (HSPGs) are complex molecules involved in the growth, invasion and metastatic properties of cancerous cells. This study analyses the alterations in the expression patterns of these molecules in right sided colorectal cancer (CRC), both metastatic and non-metastatic. Methods Twenty right sided CRCs were studied. A transcriptomic approach was used, employing qPCR to analyze both the expression of the enzymes involved in heparan sulfate (HS) chains biosynthesis, as well as the proteoglycan core proteins. Since some of these proteoglycans can also carry chondroitin sulfate (CS) chains, we include the study of the genes involved in the biosynthesis of these glycosaminoglycans. Immunohistochemical techniques were also used to analyze tissue expression of particular genes showing significant expression differences, of potential interest. Results Changes in proteoglycan core proteins differ depending on their location; those located intracellularly or in the extracellular matrix show very similar alteration patterns, while those located on the cell surface vary greatly depending on the nature of the tumor: glypicans 1, 3, 6 and betaglycan are affected in the non-metastatic tumors, whereas in the metastatic, only glypican-1 and syndecan-1 are modified, the latter showing opposing alterations in levels of RNA and of protein, suggesting post-transcriptional regulation in these tumors. Furthermore, in non-metastatic tumors, polymerization of glycosaminoglycan chains is modified, particularly affecting the synthesis of the tetrasaccharide linker and the initiation and elongation of CS chains, HS chains being less affected. Regarding the enzymes responsible for the modificaton of the HS chains, alterations were only found in non-metastatic tumors, affecting N-sulfation and the isoforms HS6ST1, HS3ST3B and HS3ST5. In contrast, synthesis of the CS chains suggests changes in epimerization and sulfation of the C4 and C2 in both types of tumor. Conclusions Right sided CRCs show alterations in the expression of HSPGs, including the expression of the cell surface core proteins, many glycosiltransferases and some enzymes that modify the HS chains depending on the metastatic nature of the tumor, resulting more affected in non-metastatic ones. However, matrix proteoglycans and enzymes involved in CS fine structure synthesis are extensively modified independetly of the presence of lymph node metastasis. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1724-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Iván Fernández-Vega
- Servicio de Patología. Hospital Universitario de Araba, Álava, 01009, Spain.
| | - Olivia García-Suárez
- Department of Morphology and Cell Biology, University of Oviedo, 33006, Oviedo, Spain.
| | - Beatriz García
- University Institute of Oncology of Asturias, Oviedo, Spain. .,Department of Functional Biology, University of Oviedo, 33006, Oviedo, Spain.
| | - Ainara Crespo
- Department of Biotechnology, Neiker-Tecnalia Arkaute, 01080, Vitoria-Gasteiz, Spain.
| | - Aurora Astudillo
- University Institute of Oncology of Asturias, Oviedo, Spain. .,Department of Pathology, Hospital, Universitario Central de Asturias, 33006, Oviedo, Spain.
| | - Luis M Quirós
- University Institute of Oncology of Asturias, Oviedo, Spain. .,Department of Functional Biology, University of Oviedo, 33006, Oviedo, Spain.
| |
Collapse
|
50
|
|