1
|
Yablecovitch D, Nadler M, Ben‐Horin S, Picard O, Yavzori M, Fudim E, Duchan MT, Sakhnini E, Lang A, Lahav M, Saker T, Neuman S, Selinger L, Freitz B, Dvir R, Raitses‐Gurevich M, Golan T, Levy I, Laish I. Serum matrix metalloproteinase-7, Syndecan-1, and CA 19-9 as a biomarker panel for diagnosis of pancreatic ductal adenocarcinoma. Cancer Med 2024; 13:e70144. [PMID: 39263943 PMCID: PMC11391268 DOI: 10.1002/cam4.70144] [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: 05/17/2024] [Revised: 07/29/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024] Open
Abstract
AIMS AND BACKGROUND Matrix metalloproteinase-7 (MMP-7) and Syndecan-1 (SDC1) are involved in multiple functions during tumorigenesis. We aimed to evaluate the diagnostic and prognostic performance of these serum proteins, as potential biomarkers, in patients with pancreatic ductal adenocarcinoma (PDAC) and benign pancreatic cysts. METHODS In this case-control study, patients with newly diagnosed PDAC (N = 121) were compared with the benign cyst (N = 66) and healthy control (N = 48) groups. Serum MMP-7 and SDC1 were measured by ELISA. The diagnostic accuracy of their levels for diagnosing PDAC and pancreatic cysts was computed, and their association with survival outcomes was evaluated. RESULTS MMP-7 median serum levels were significantly elevated in the PDAC (7.3 ng/mL) and cyst groups (3.7 ng/mL) compared with controls (2.9 ng/mL) (p < 0.001 and 0.02, respectively), and also between the PDAC and cyst groups (p < 0.001), while SDC1 median serum levels were significantly elevated in PDAC (43.3 ng/mL) compared with either cysts (30.1 ng/mL, p < 0.001) or controls (31.2 ng/mL, p < 0.001). The receiver operating characteristic curve analysis area under the curve in PDAC versus controls was 0.90 and 0.78 for MMP-7 and SDC1, respectively, while it was 1.0 for the combination of the two and CA 19-9 (p < 0.001). The combination of the three biomarkers had a perfect sensitivity (100%). CONCLUSIONS Due to its high sensitivity, this biomarker panel has the potential to rule out PDAC in suspected cases.
Collapse
Affiliation(s)
- Doron Yablecovitch
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Moshe Nadler
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Shomron Ben‐Horin
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Orit Picard
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Miri Yavzori
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Ella Fudim
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Moran Tardio Duchan
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Emad Sakhnini
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Alon Lang
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Maor Lahav
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Talia Saker
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
- Shalvata Mental Health CenterHod HasharonIsrael
| | - Sandra Neuman
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Limor Selinger
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Biana Freitz
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Revital Dvir
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Maria Raitses‐Gurevich
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
- Department of OncologyChaim Sheba Medical CenterTel HashomerIsrael
| | - Talia Golan
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
- Department of OncologyChaim Sheba Medical CenterTel HashomerIsrael
| | - Idan Levy
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Ido Laish
- Gastroenterology InstituteChaim Sheba Medical CenterTel HashomerIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
2
|
Kim S, Yang H, Cho S, Jang Y, Han IO, Oh ES. Correlation of syndecan gene amplification with metastatic potential and clinical outcomes in carcinomas. Am J Physiol Cell Physiol 2024; 327:C380-C386. [PMID: 38953842 DOI: 10.1152/ajpcell.00270.2024] [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: 04/22/2024] [Revised: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024]
Abstract
Cell surface receptors play crucial roles in cellular responses to extracellular ligands, helping to modulate the functions of a cell based on information coming from outside the cell. Syndecan refers to a family of cell adhesion receptors that regulate both extracellular and cytosolic events. Alteration of syndecan expression disrupts regulatory mechanisms in a cell type-specific fashion, often leading to serious diseases, notably cancer. Given the multifaceted functions and distinct tissue distributions of syndecan, it will be important to unravel the gene-level intricacies of syndecan expression and thereby further understand its involvement in various carcinogenic processes. Although accumulating evidence indicates that the protein expression patterns of syndecan family members are significantly altered in cancer cells, the underlying gene-level mechanisms remain largely unknown. This review endeavors to explore syndecan gene expression levels across different cancer types by scrutinizing extensive cancer genome datasets using tools such as cBioPortal. Our analysis unveils that somatic mutations in SDC genes are rare occurrences, whereas copy number alterations are frequently observed across diverse cancers, particularly in SDC2 and SDC4. Notably, amplifications of SDC2 and SDC4 correlate with heightened metastatic potential and dismal prognosis. This underscores the recurrent nature of SDC2 and SDC4 amplifications during carcinogenesis and sheds light on their role in promoting cancer activity through augmented protein expression. The identification of these amplifications not only enriches our understanding of carcinogenic mechanisms but also hints at the potential therapeutic avenue of targeting SDC2 and SDC4 to curb cancer cell proliferation and metastasis.
Collapse
Affiliation(s)
- Sewoon Kim
- Institute of Sensor Technology, Easytem Co., Ltd., Seoul, Republic of Korea
| | - Hyeonju Yang
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Subin Cho
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Yunjung Jang
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Inn-Oc Han
- Department of Biomedical Science, Program in Biomedical Science and Engineering, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Eok-Soo Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Pinheiro AHG, Pereira BDO, Silva LSD, de Melo FTC, de Souza ACCB, Leal VSG, de Figueiredo PBB, Neto JFA, dos Santos MC, de Queiroz NNM, Felício KM, Ribeiro-dos-Santos Â, Felício JS, Cavalcante GC. Downregulation of hsa-miR-100-5p May Be a Protective Factor in the Early Stages of Nephropathy in Type 1 Diabetes Mellitus. Int J Mol Sci 2024; 25:5663. [PMID: 38891851 PMCID: PMC11171847 DOI: 10.3390/ijms25115663] [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: 04/16/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Type 1 Diabetes Mellitus (T1DM) can generate severe complications, such as Diabetic Kidney Disease (DKD) or Diabetic Nephropathy (DN), with it emerging as the leading cause of terminal (end-stage) renal disease all over the world. For T1DM, the clinical evaluation of DKD uses markers like the Glomerular Filtration Rate (GFR) and the Urinary Albumin Excretion (UAE). However, early diagnosis of DKD is still a challenge. For this reason, investigating molecular markers, such as microRNAs (miRNAs), offers a promising perspective to an early diagnosis, highlighting the stability and the ability to reflect incipient molecular manifestations. Thus, here we investigated four miRNAs (hsa-let-7i-5p, hsa-miR-143-3p, hsa-miR-501-3p, and hsa-miR-100-5p) regarding nephropathy in patients with T1DM, considering the albuminuria (micro and macro) as a standard to evaluate the groups. As a result, we found a reduced expression of miR-100-5p in patients with MIC, indicating a protective role in nephropathy. Beyond that, expression levels between the groups (Non vs. UAE) were not significant when comparing the miRNAs miR-501-3p and miR-143-3p. Finally, miR-143-3p and miR-100-5p were linked to some target genes such as AKT1, MMP13, and IGF1R, that are connected to signal pathways and cellular metabolism.
Collapse
Affiliation(s)
- Andrey Henrique Gama Pinheiro
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Pará, Belém 66075-110, PA, Brazil; (A.H.G.P.); (B.d.O.P.); (Â.R.-d.-S.)
| | - Beatriz de Oliveira Pereira
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Pará, Belém 66075-110, PA, Brazil; (A.H.G.P.); (B.d.O.P.); (Â.R.-d.-S.)
| | - Lilian Souza D’Albuquerque Silva
- Endocrinology and Metabology/Diabetes Unit, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.S.D.S.); (F.T.C.d.M.); (A.C.C.B.d.S.); (V.S.G.L.); (P.B.B.d.F.); (J.F.A.N.); (M.C.d.S.); (N.N.M.d.Q.); (K.M.F.)
| | - Franciane T. Cunha de Melo
- Endocrinology and Metabology/Diabetes Unit, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.S.D.S.); (F.T.C.d.M.); (A.C.C.B.d.S.); (V.S.G.L.); (P.B.B.d.F.); (J.F.A.N.); (M.C.d.S.); (N.N.M.d.Q.); (K.M.F.)
| | - Ana Carolina C. Braga de Souza
- Endocrinology and Metabology/Diabetes Unit, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.S.D.S.); (F.T.C.d.M.); (A.C.C.B.d.S.); (V.S.G.L.); (P.B.B.d.F.); (J.F.A.N.); (M.C.d.S.); (N.N.M.d.Q.); (K.M.F.)
| | - Valéria S. Galvão Leal
- Endocrinology and Metabology/Diabetes Unit, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.S.D.S.); (F.T.C.d.M.); (A.C.C.B.d.S.); (V.S.G.L.); (P.B.B.d.F.); (J.F.A.N.); (M.C.d.S.); (N.N.M.d.Q.); (K.M.F.)
| | - Priscila B. Barbosa de Figueiredo
- Endocrinology and Metabology/Diabetes Unit, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.S.D.S.); (F.T.C.d.M.); (A.C.C.B.d.S.); (V.S.G.L.); (P.B.B.d.F.); (J.F.A.N.); (M.C.d.S.); (N.N.M.d.Q.); (K.M.F.)
| | - João F. Abrahão Neto
- Endocrinology and Metabology/Diabetes Unit, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.S.D.S.); (F.T.C.d.M.); (A.C.C.B.d.S.); (V.S.G.L.); (P.B.B.d.F.); (J.F.A.N.); (M.C.d.S.); (N.N.M.d.Q.); (K.M.F.)
| | - Marcia Costa dos Santos
- Endocrinology and Metabology/Diabetes Unit, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.S.D.S.); (F.T.C.d.M.); (A.C.C.B.d.S.); (V.S.G.L.); (P.B.B.d.F.); (J.F.A.N.); (M.C.d.S.); (N.N.M.d.Q.); (K.M.F.)
| | - Natércia Neves Marques de Queiroz
- Endocrinology and Metabology/Diabetes Unit, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.S.D.S.); (F.T.C.d.M.); (A.C.C.B.d.S.); (V.S.G.L.); (P.B.B.d.F.); (J.F.A.N.); (M.C.d.S.); (N.N.M.d.Q.); (K.M.F.)
| | - Karem Miléo Felício
- Endocrinology and Metabology/Diabetes Unit, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.S.D.S.); (F.T.C.d.M.); (A.C.C.B.d.S.); (V.S.G.L.); (P.B.B.d.F.); (J.F.A.N.); (M.C.d.S.); (N.N.M.d.Q.); (K.M.F.)
| | - Ândrea Ribeiro-dos-Santos
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Pará, Belém 66075-110, PA, Brazil; (A.H.G.P.); (B.d.O.P.); (Â.R.-d.-S.)
| | - João Soares Felício
- Endocrinology and Metabology/Diabetes Unit, João de Barros Barreto University Hospital, Federal University of Pará, Belém 66075-110, PA, Brazil; (L.S.D.S.); (F.T.C.d.M.); (A.C.C.B.d.S.); (V.S.G.L.); (P.B.B.d.F.); (J.F.A.N.); (M.C.d.S.); (N.N.M.d.Q.); (K.M.F.)
| | - Giovanna C. Cavalcante
- Laboratory of Human and Medical Genetics, Graduate Program in Genetics and Molecular Biology, Federal University of Pará, Belém 66075-110, PA, Brazil; (A.H.G.P.); (B.d.O.P.); (Â.R.-d.-S.)
| |
Collapse
|
4
|
Riccardi F, Tangredi C, Dal Bo M, Toffoli G. Targeted therapy for multiple myeloma: an overview on CD138-based strategies. Front Oncol 2024; 14:1370854. [PMID: 38655136 PMCID: PMC11035824 DOI: 10.3389/fonc.2024.1370854] [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: 01/15/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024] Open
Abstract
Multiple myeloma (MM) is an incurable hematological disease characterized by the uncontrolled growth of plasma cells primarily in the bone marrow. Although its treatment consists of the administration of combined therapy regimens mainly based on immunomodulators and proteosome inhibitors, MM remains incurable, and most patients suffer from relapsed/refractory disease with poor prognosis and survival. The robust results achieved by immunotherapy targeting MM-associated antigens CD38 and CD319 (also known as SLAMF7) have drawn attention to the development of new immune-based strategies and different innovative compounds in the treatment of MM, including new monoclonal antibodies, antibody-drug conjugates, recombinant proteins, synthetic peptides, and adaptive cellular therapies. In this context, Syndecan1 (CD138 or SDC1), a transmembrane heparan sulfate proteoglycan that is upregulated in malignant plasma cells, has gained increasing attention in the panorama of MM target antigens, since its key role in MM tumorigenesis, progression and aggressiveness has been largely reported. Here, our aim is to provide an overview of the most important aspects of MM disease and to investigate the molecular functions of CD138 in physiologic and malignant cell states. In addition, we will shed light on the CD138-based therapeutic approaches currently being tested in preclinical and/or clinical phases in MM and discuss their properties, mechanisms of action and clinical applications.
Collapse
Affiliation(s)
- Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Carmela Tangredi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
5
|
Festari MF, Jara E, Costa M, Iriarte A, Freire T. Truncated O-glycosylation in metastatic triple-negative breast cancer reveals a gene expression signature associated with extracellular matrix and proteolysis. Sci Rep 2024; 14:1809. [PMID: 38245559 PMCID: PMC10799929 DOI: 10.1038/s41598-024-52204-2] [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: 11/08/2023] [Accepted: 01/16/2024] [Indexed: 01/22/2024] Open
Abstract
Breast cancer (BC) is the leading cause of death by cancer in women worldwide. Triple-negative (TN) BC constitutes aggressive and highly metastatic tumors associated with shorter overall survival of patients compared to other BC subtypes. The Tn antigen, a glycoconjugated structure resulting from an incomplete O-glycosylation process, is highly expressed in different adenocarcinomas, including BC. It also favors cancer growth, immunoregulation, and metastasis in TNBC. This work describes the differentially expressed genes (DEGs) associated with BC aggressiveness and metastasis in an incomplete O-glycosylated TNBC cell model. We studied the transcriptome of a TNBC model constituted by the metastatic murine 4T1 cell line that overexpresses the Tn antigen due to a mutation in one of the steps of the O-glycosylation pathway. We analyzed and compared the results with the parental wild-type cell line and with a Tn-negative cell clone that was poorly metastatic and less aggressive than the 4T1 parental cell line. To gain insight into the generated expression data, we performed a gene set analysis. Biological processes associated with cancer development and metastasis, immune evasion, and leukocyte recruitment were highly enriched among functional terms of DEGs. Furthermore, different highly O-glycosylated protein-coding genes, such as mmp9, ecm1 and ankyrin-2, were upregulated in 4T1/Tn+ tumor cells. The altered biological processes and DEGs that promote tumor growth, invasion and immunomodulation might explain the aggressive properties of 4T1/Tn+ tumor cells. These results support the hypothesis that incomplete O-glycosylation that leads to the expression of the Tn antigen, which might regulate activity or interaction of different molecules, promotes cancer development and immunoregulation.
Collapse
Affiliation(s)
- María Florencia Festari
- Laboratorio de Inmunomodulación y Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, 11800, Montevideo, Uruguay
| | - Eugenio Jara
- Unidad de Genética y Mejora Animal, Departamento de Producción Animal, Facultad de Veterinaria, Universidad de la República, Montevideo, Uruguay
| | - Monique Costa
- Laboratorio de Inmunomodulación y Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, 11800, Montevideo, Uruguay
| | - Andrés Iriarte
- Laboratorio de Biología Computacional, Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Dr. Alfredo Navarro 3051, 11600, Montevideo, Uruguay.
| | - Teresa Freire
- Laboratorio de Inmunomodulación y Vacunas, Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Gral. Flores 2125, 11800, Montevideo, Uruguay.
| |
Collapse
|
6
|
Wang X, Zhang Q, Ren Y, Liu C, Gao H. Research Progress on Extracellular Matrix Involved in the Development of Preeclampsia. Curr Protein Pept Sci 2024; 25:527-538. [PMID: 38561606 DOI: 10.2174/0113892037284176240302052521] [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: 09/27/2023] [Revised: 01/21/2024] [Accepted: 02/14/2024] [Indexed: 04/04/2024]
Abstract
Preeclampsia (PE) is a serious pregnancy complication, and its primary clinical manifestations are gestational hypertension and proteinuria. Trophoblasts are responsible for the basic functions of the placenta during placental development; recent studies have revealed that placental "shallow implantation" caused by the decreased invasiveness of placental trophoblasts plays a crucial role in PE pathogenesis. The interaction between the cells and the extracellular matrix (ECM) plays a crucial role in trophoblast proliferation, differentiation, and invasion. Abnormal ECM function can result in insufficient migration and invasion of placental trophoblasts, thus participating in PE. This article summarizes the recent studies on the involvement of ECM components, including small leucine-rich proteoglycans, syndecans, glypicans, laminins, fibronectin, collagen, and hyaluronic acid, in the development of PE. ECM plays various roles in PE development, most notably by controlling the activities of trophoblasts. The ECM is structurally stable and can serve as a biological diagnostic marker and therapeutic target for PE.
Collapse
Affiliation(s)
- Xin Wang
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
- College of Pharmacy, Jining Medical University, Rizhao, Shandong, China
| | - Qi Zhang
- Department of Pharmacy, Shandong First Medical University, Jinan, Shandong, China
| | - Yi Ren
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Chao Liu
- College of Pharmacy, Jining Medical University, Rizhao, Shandong, China
| | - Huijie Gao
- College of Pharmacy, Jining Medical University, Rizhao, Shandong, China
| |
Collapse
|
7
|
Hu H, He B, He M, Tao H, Li B. A glycosylation-related signature predicts survival in pancreatic cancer. Aging (Albany NY) 2023; 15:13710-13737. [PMID: 38048216 PMCID: PMC10756102 DOI: 10.18632/aging.205258] [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: 02/24/2023] [Accepted: 10/19/2023] [Indexed: 12/06/2023]
Abstract
BACKGROUND Tumor initiation and progression are closely associated with glycosylation. However, glycosylated molecules have not been the subject of extensive studies as prognostic markers for pancreatic cancer. The objectives of this study were to identify glycosylation-related genes in pancreatic cancer and use them to construct reliable prognostic models. MATERIALS AND METHODS The Cancer Genome Atlas and Gene Expression Omnibus databases were used to assess the differential expression of glycosylation-related genes; four clusters were identified based on consistent clustering analysis. Kaplan-Meier analyses identified three glycosylation-related genes associated with overall survival. LASSO analysis was then performed on The Cancer Genome Atlas and International Cancer Genome Consortium databases to identify glycosylation-related signatures. We identified 12 GRGs differently expressed in pancreatic cancer and selected three genes (SEL1L, TUBA1C, and SDC1) to build a prognostic model. Thereafter, patients were divided into high and low-risk groups. Eventually, we performed Quantitative real-time PCR (qRT-PCR) to validate the signature. RESULTS Clinical outcomes were significantly poorer in the high-risk group than in the low-risk group. There were also significant correlations between the high-risk group and several risk factors, including no-smoking history, drinking history, radiotherapy history, and lower tumor grade. Furthermore, the high-risk group had a higher proportion of immune cells. Eventually, three glycosylation-related genes were validated in human PC cell lines. CONCLUSION This study identified the glycosylation-related signature for pancreatic cancer. It is an effective predictor of survival and can guide treatment decisions.
Collapse
Affiliation(s)
- Huidong Hu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Bingsheng He
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Mingang He
- Department of Gastrointestinal Surgery, Shandong Tumor Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Hengmin Tao
- Department of Head and Neck Radiotherapy, Shandong Provincial ENT Hospital, Shandong University, Jinan 250117, China
| | - Baosheng Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
8
|
Ahn S, Sharma U, Kasuba KC, Strohmeyer N, Müller DJ. Engineered Biomimetic Fibrillar Fibronectin Matrices Regulate Cell Adhesion Initiation, Migration, and Proliferation via α5β1 Integrin and Syndecan-4 Crosstalk. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300812. [PMID: 37357136 PMCID: PMC10460904 DOI: 10.1002/advs.202300812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/24/2023] [Indexed: 06/27/2023]
Abstract
Cells regulate adhesion to the fibrillar extracellular matrix (ECM) of which fibronectin is an essential component. However, most studies characterize cell adhesion to globular fibronectin substrates at time scales long after cells polarize and migrate. To overcome this limitation, a simple and scalable method to engineer biomimetic 3D fibrillar fibronectin matrices is introduced and how they are sensed by fibroblasts from the onset of attachment is characterized. Compared to globular fibronectin substrates, fibroblasts accelerate adhesion initiation and strengthening within seconds to fibrillar fibronectin matrices via α5β1 integrin and syndecan-4. This regulation, which additionally accelerates on stiffened fibrillar matrices, involves actin polymerization, actomyosin contraction, and the cytoplasmic proteins paxillin, focal adhesion kinase, and phosphoinositide 3-kinase. Furthermore, this immediate sensing and adhesion of fibroblast to fibrillar fibronectin guides migration speed, persistency, and proliferation range from hours to weeks. The findings highlight that fibrillar fibronectin matrices, compared to widely-used globular fibronectin, trigger short- and long-term cell decisions very differently and urge the use of such matrices to better understand in vivo interactions of cells and ECMs. The engineered fibronectin matrices, which can be printed onto non-biological surfaces without loss of function, open avenues for various cell biological, tissue engineering and medical applications.
Collapse
Affiliation(s)
- Seungkuk Ahn
- Department of Biosystems Science and EngineeringEidgenössische Technische Hochschule (ETH) ZurichBasel4058Switzerland
| | - Upnishad Sharma
- Department of Biosystems Science and EngineeringEidgenössische Technische Hochschule (ETH) ZurichBasel4058Switzerland
| | - Krishna Chaitanya Kasuba
- Department of Biosystems Science and EngineeringEidgenössische Technische Hochschule (ETH) ZurichBasel4058Switzerland
| | - Nico Strohmeyer
- Department of Biosystems Science and EngineeringEidgenössische Technische Hochschule (ETH) ZurichBasel4058Switzerland
| | - Daniel J. Müller
- Department of Biosystems Science and EngineeringEidgenössische Technische Hochschule (ETH) ZurichBasel4058Switzerland
| |
Collapse
|
9
|
Ricard-Blum S, Couchman JR. Conformations, interactions and functions of intrinsically disordered syndecans. Biochem Soc Trans 2023:BST20221085. [PMID: 37334846 DOI: 10.1042/bst20221085] [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: 02/19/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/21/2023]
Abstract
Syndecans are transmembrane heparan sulfate proteoglycans present on most mammalian cell surfaces. They have a long evolutionary history, a single syndecan gene being expressed in bilaterian invertebrates. Syndecans have attracted interest because of their potential roles in development and disease, including vascular diseases, inflammation and various cancers. Recent structural data is providing important insights into their functions, which are complex, involving both intrinsic signaling through cytoplasmic binding partners and co-operative mechanisms where syndecans form a signaling nexus with other receptors such as integrins and tyrosine kinase growth factor receptors. While the cytoplasmic domain of syndecan-4 has a well-defined dimeric structure, the syndecan ectodomains are intrinsically disordered, which is linked to a capacity to interact with multiple partners. However, it remains to fully establish the impact of glycanation and partner proteins on syndecan core protein conformations. Genetic models indicate that a conserved property of syndecans links the cytoskeleton to calcium channels of the transient receptor potential class, compatible with roles as mechanosensors. In turn, syndecans influence actin cytoskeleton organization to impact motility, adhesion and the extracellular matrix environment. Syndecan clustering with other cell surface receptors into signaling microdomains has relevance to tissue differentiation in development, for example in stem cells, but also in disease where syndecan expression can be markedly up-regulated. Since syndecans have potential as diagnostic and prognostic markers as well as possible targets in some forms of cancer, it remains important to unravel structure/function relationships in the four mammalian syndecans.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- ICBMS, UMR 5246 CNRS, Universite Claude Bernard Lyon 1, F-69622 Villeurbanne, France
| | - John R Couchman
- Biotech Research & Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
10
|
Oto J, Le QK, Schäfer SD, Kiesel L, Marí-Alexandre J, Gilabert-Estellés J, Medina P, Götte M. Role of Syndecans in Ovarian Cancer: New Diagnostic and Prognostic Biomarkers and Potential Therapeutic Targets. Cancers (Basel) 2023; 15:3125. [PMID: 37370735 DOI: 10.3390/cancers15123125] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/22/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Ovarian cancer (OC) is the eighth cancer both in prevalence and mortality in women and represents the deadliest female reproductive cancer. Due to generally vague symptoms, OC is frequently diagnosed only at a late and advanced stage, resulting in high mortality. The tumor extracellular matrix and cellular matrix receptors play a key role in the pathogenesis of tumor progression. Syndecans are a family of four transmembrane heparan sulfate proteoglycans (PG), including syndecan-1, -2, -3, and -4, which are dysregulated in a myriad of cancers, including OC. Many clinicopathological studies suggest that these proteins are promising diagnostic and prognostic biomarkers for OC. Furthermore, functions of the syndecan family in the regulation of cellular processes make it an interesting pharmacological target for anticancer therapies.
Collapse
Affiliation(s)
- Julia Oto
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, 46026 Valencia, Spain
| | - Quang-Khoi Le
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany
| | - Sebastian D Schäfer
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany
| | - Josep Marí-Alexandre
- Research Laboratory in Biomarkers in Reproduction, Gynaecology and Obstetrics, Fundación Hospital General Universitario de Valencia, 46014 Valencia, Spain
- Department of Pathology, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain
| | - Juan Gilabert-Estellés
- Research Laboratory in Biomarkers in Reproduction, Gynaecology and Obstetrics, Fundación Hospital General Universitario de Valencia, 46014 Valencia, Spain
- Department of Gynecology and Obstetrics, Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain
- Department of Paediatrics, Obstetrics and Gynecology, University of Valencia, 46010 Valencia, Spain
| | - Pilar Medina
- Haemostasis, Thrombosis, Arteriosclerosis and Vascular Biology Research Group, Medical Research Institute Hospital La Fe, 46026 Valencia, Spain
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, 48149 Münster, Germany
| |
Collapse
|
11
|
Raskov H, Gaggar S, Tajik A, Orhan A, Gögenur I. The Matrix Reloaded-The Role of the Extracellular Matrix in Cancer. Cancers (Basel) 2023; 15:2057. [PMID: 37046716 PMCID: PMC10093330 DOI: 10.3390/cancers15072057] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
As the core component of all organs, the extracellular matrix (ECM) is an interlocking macromolecular meshwork of proteins, glycoproteins, and proteoglycans that provides mechanical support to cells and tissues. In cancer, the ECM can be remodelled in response to environmental cues, and it controls a plethora of cellular functions, including metabolism, cell polarity, migration, and proliferation, to sustain and support oncogenesis. The biophysical and biochemical properties of the ECM, such as its structural arrangement and being a reservoir for bioactive molecules, control several intra- and intercellular signalling pathways and induce cytoskeletal changes that alter cell shapes, behaviour, and viability. Desmoplasia is a major component of solid tumours. The abnormal deposition and composition of the tumour matrix lead to biochemical and biomechanical alterations that determine disease development and resistance to treatment. This review summarises the complex roles of ECM in cancer and highlights the possible therapeutic targets and how to potentially remodel the dysregulated ECM in the future. Furthering our understanding of the ECM in cancer is important as the modification of the ECM will probably become an important tool in the characterisation of individual tumours and personalised treatment options.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Shruti Gaggar
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Asma Tajik
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Oncology, Zealand University Hospital, 4000 Roskilde, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
12
|
Kyriakopoulou K, Piperigkou Z, Tzaferi K, Karamanos NK. Trends in extracellular matrix biology. Mol Biol Rep 2023; 50:853-863. [PMID: 36342580 PMCID: PMC9884264 DOI: 10.1007/s11033-022-07931-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/06/2022] [Indexed: 11/09/2022]
Abstract
Extracellular matrixes (ECMs) are intricate 3-dimensional macromolecular networks of unique architectures with regulatory roles in cell morphology and functionality. As a dynamic native biomaterial, ECM undergoes constant but tightly controlled remodeling that is crucial for the maintenance of normal cellular behavior. Under pathological conditions like cancer, ECM remodeling ceases to be subjected to control resulting in disease initiation and progression. ECM is comprised of a staggering number of molecules that interact not only with one another, but also with neighboring cells via cell surface receptors. Such interactions, too many to tally, are of paramount importance for the identification of novel disease biomarkers and more personalized therapeutic intervention. Recent advances in big data analytics have allowed the development of online databases where researchers can take advantage of a stochastic evaluation of all the possible interactions and narrow them down to only those of interest for their study, respectively. This novel approach addresses the limitations that currently exist in studies, expands our understanding on ECM interactions, and has the potential to advance the development of targeted therapies. In this article we present the current trends in ECM biology research and highlight its importance in tissue integrity, the main interaction networks, ECM-mediated cell functional properties and issues related to pharmacological targeting.
Collapse
Affiliation(s)
- Konstantina Kyriakopoulou
- Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04, Patras, Greece
| | - Zoi Piperigkou
- Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04, Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH), Institute of Chemical Engineering Sciences (ICE-HT), 261 10, Patras, Greece
| | - Kyriaki Tzaferi
- Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04, Patras, Greece
| | - Nikos K Karamanos
- Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 265 04, Patras, Greece.
- Foundation for Research and Technology-Hellas (FORTH), Institute of Chemical Engineering Sciences (ICE-HT), 261 10, Patras, Greece.
| |
Collapse
|
13
|
Schüler SC, Liu Y, Dumontier S, Grandbois M, Le Moal E, Cornelison DDW, Bentzinger CF. Extracellular matrix: Brick and mortar in the skeletal muscle stem cell niche. Front Cell Dev Biol 2022; 10:1056523. [PMID: 36523505 PMCID: PMC9745096 DOI: 10.3389/fcell.2022.1056523] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/03/2022] [Indexed: 11/30/2022] Open
Abstract
The extracellular matrix (ECM) is an interconnected macromolecular scaffold occupying the space between cells. Amongst other functions, the ECM provides structural support to tissues and serves as a microenvironmental niche that conveys regulatory signals to cells. Cell-matrix adhesions, which link the ECM to the cytoskeleton, are dynamic multi-protein complexes containing surface receptors and intracellular effectors that control various downstream pathways. In skeletal muscle, the most abundant tissue of the body, each individual muscle fiber and its associated muscle stem cells (MuSCs) are surrounded by a layer of ECM referred to as the basal lamina. The core scaffold of the basal lamina consists of self-assembling polymeric laminins and a network of collagens that tether proteoglycans, which provide lateral crosslinking, establish collateral associations with cell surface receptors, and serve as a sink and reservoir for growth factors. Skeletal muscle also contains the fibrillar collagenous interstitial ECM that plays an important role in determining tissue elasticity, connects the basal laminae to each other, and contains matrix secreting mesenchymal fibroblast-like cell types and blood vessels. During skeletal muscle regeneration fibroblast-like cell populations expand and contribute to the transitional fibronectin-rich regenerative matrix that instructs angiogenesis and MuSC function. Here, we provide a comprehensive overview of the role of the skeletal muscle ECM in health and disease and outline its role in orchestrating tissue regeneration and MuSC function.
Collapse
Affiliation(s)
- Svenja C. Schüler
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Yuguo Liu
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Simon Dumontier
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Michel Grandbois
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Emmeran Le Moal
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - DDW Cornelison
- Division of Biological Sciences Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - C. Florian Bentzinger
- Département de Pharmacologie-Physiologie, Faculté de Médecine et des Sciences de la Santé, Institut de Pharmacologie de Sherbrooke, Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
14
|
Deb G, Cicala A, Papadas A, Asimakopoulos F. Matrix proteoglycans in tumor inflammation and immunity. Am J Physiol Cell Physiol 2022; 323:C678-C693. [PMID: 35876288 PMCID: PMC9448345 DOI: 10.1152/ajpcell.00023.2022] [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: 01/20/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 11/22/2022]
Abstract
Cancer immunoediting progresses through elimination, equilibrium, and escape. Each of these phases is characterized by breaching, remodeling, and rebuilding tissue planes and structural barriers that engage extracellular matrix (ECM) components, in particular matrix proteoglycans. Some of the signals emanating from matrix proteoglycan remodeling are readily co-opted by the growing tumor to sustain an environment of tumor-promoting and immune-suppressive inflammation. Yet other matrix-derived cues can be viewed as part of a homeostatic response by the host, aiming to eliminate the tumor and restore tissue integrity. These latter signals may be harnessed for therapeutic purposes to tip the polarity of the tumor immune milieu toward anticancer immunity. In this review, we attempt to showcase the importance and complexity of matrix proteoglycan signaling in both cancer-restraining and cancer-promoting inflammation. We propose that the era of matrix diagnostics and therapeutics for cancer is fast approaching the clinic.
Collapse
Affiliation(s)
- Gauri Deb
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, California
- Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, California
| | - Alexander Cicala
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, California
- Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, California
| | - Athanasios Papadas
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, California
- Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, California
| | - Fotis Asimakopoulos
- Division of Blood and Marrow Transplantation, Department of Medicine, University of California, San Diego (UCSD), La Jolla, California
- Moores Cancer Center, University of California, San Diego (UCSD), La Jolla, California
| |
Collapse
|
15
|
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: 0.5] [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
|
16
|
Kines RC, Schiller JT. Harnessing Human Papillomavirus' Natural Tropism to Target Tumors. Viruses 2022; 14:1656. [PMID: 36016277 PMCID: PMC9413966 DOI: 10.3390/v14081656] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023] Open
Abstract
Human papillomaviruses (HPV) are small non-enveloped DNA tumor viruses established as the primary etiological agent for the development of cervical cancer. Decades of research have elucidated HPV's primary attachment factor to be heparan sulfate proteoglycans (HSPG). Importantly, wounding and exposure of the epithelial basement membrane was found to be pivotal for efficient attachment and infection of HPV in vivo. Sulfation patterns on HSPG's become modified at the site of wounds as they serve an important role promoting tissue healing, cell proliferation and neovascularization and it is these modifications recognized by HPV. Analogous HSPG modification patterns can be found on tumor cells as they too require the aforementioned processes to grow and metastasize. Although targeting tumor associated HSPG is not a novel concept, the use of HPV to target and treat tumors has only been realized in recent years. The work herein describes how decades of basic HPV research has culminated in the rational design of an HPV-based virus-like infrared light activated dye conjugate for the treatment of choroidal melanoma.
Collapse
Affiliation(s)
| | - John T. Schiller
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA;
| |
Collapse
|
17
|
Yang Z, Chen S, Ying H, Yao W. Targeting syndecan-1: new opportunities in cancer therapy. Am J Physiol Cell Physiol 2022; 323:C29-C45. [PMID: 35584326 PMCID: PMC9236862 DOI: 10.1152/ajpcell.00024.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022]
Abstract
Syndecan-1 (SDC1, CD138) is one of the heparan sulfate proteoglycans and is essential for maintaining normal cell morphology, interacting with the extracellular and intracellular protein repertoire, as well as mediating signaling transduction upon environmental stimuli. The critical role of SDC1 in promoting tumorigenesis and metastasis has been increasingly recognized in various cancer types, implying a promising potential of utilizing SDC1 as a novel target for cancer therapy. This review summarizes the current knowledge on SDC1 structure and functions, including its role in tumor biology. We also discuss the highlights and limitations of current SDC1-targeted therapies as well as the obstacles in developing new therapeutic methods, offering our perspective on the future directions to target SDC1 for cancer treatment.
Collapse
Affiliation(s)
- Zecheng Yang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shuaitong Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wantong Yao
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
18
|
Reszegi A, Tátrai P, Regős E, Kovalszky I, Baghy K. Syndecan-1 in liver pathophysiology. Am J Physiol Cell Physiol 2022; 323:C289-C294. [PMID: 35704700 DOI: 10.1152/ajpcell.00039.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Syndecan-1 is a heparan sulfate/chondroitin sulfate proteoglycan (PG) of the cell surface and the extracellular matrix, which regulates a broad spectrum of physiological and pathological processes such as cell proliferation, migration, inflammation, matrix remodeling, wound healing, or tumorigenesis. Syndecan-1 represents the major PG of the liver, expressed by hepatocytes and cholangiocytes, and its elevated expression is a characteristic feature of liver diseases. The highest syndecan-1 expression is found in liver cirrhosis and in hepatocellular carcinoma (HCC) developed in cirrhotic livers. In addition, as being a hepatitis C receptor, hepatitis C virus (HCV) infected livers produce extremely large amounts of syndecan-1. The serum levels of the cleaved (shedded) extracellular domain has clinical significance, as its increased concentration reflects on poor prognosis in cirrhosis as well as in cancer. In vivo experiments confirmed that syndecan-1 protects against early stages of fibrogenesis mainly by enhanced clearance of transforming growth factor beta (TGFβ1) and thrombospondin-1 via circulation, and against hepatocarcinogenesis by interfering with several signaling pathways and enhancing cell cycle blockade. In addition, syndecan-1 is capable to hinder lipid metabolism and ribosomal biogenesis in induced cancer models.. These observations together with its participation in the uptake of viruses (e.g. HCV, SARS-CoV-2) indicate that syndecan-1 is a central player in liver pathologies.
Collapse
Affiliation(s)
- Andrea Reszegi
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | | | - Eszter Regős
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ilona Kovalszky
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Kornelia Baghy
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
19
|
Yablecovitch D, Ben-Horin S, Picard O, Yavzori M, Fudim E, Nadler M, Levy I, Sakhnini E, Lang A, Engel T, Lahav M, Saker T, Neuman S, Selinger L, Dvir R, Raitses-Gurevich M, Golan T, Laish I. Serum Syndecan-1: A Novel Biomarker for Pancreatic Ductal Adenocarcinoma. Clin Transl Gastroenterol 2022; 13:e00473. [PMID: 35297817 PMCID: PMC9132524 DOI: 10.14309/ctg.0000000000000473] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 02/01/2022] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Syndecan-1 (SDC1) has multiple functions in tumorigenesis in general and specifically in pancreatic cancer. We aimed to evaluate SDC1 as a diagnostic and prognostic biomarker in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS In this case-control study, patients newly diagnosed with a biopsy-proven PDAC were enrolled alongside healthy individuals in a derivation-validation cohort design. Serum SDC1 was measured by enzyme-linked immunoassay. The diagnostic accuracy of SDC1 levels for diagnosing PDAC was computed. A unified cohort enriched with additional early-stage patients with PDAC was used to evaluate the association of SDC1 with survival outcomes and patient characteristics. RESULTS In the derivation cohort, serum SDC1 levels were significantly higher in patients with PDAC (n = 39) compared with healthy controls (n = 20) (40.1 ng/mL, interquartile range 29.8-95.3 vs 25.6 ng/mL, interquartile range 17.1-29.8, respectively; P < 0.001). The receiver operating characteristic analysis area under the curve was 0.847 (95% confidence interval 0.747-0.947, P < 0.001). These results were replicated in a separate age-matched validation cohort (n = 38 PDAC, n = 38 controls; area under the curve 0.844, 95% confidence interval 0.757-0.932, P < 0.001). In the combined-enriched PDAC cohort (n = 110), using a cutoff of 35 ng/mL, the median overall 5-year survival between patients below and above this cutoff was not significantly different, although a trend for better survival after 1 year was found in the lower level group (P = 0.06). There were 12 of the 110 patients with PDAC (11%) who had normal CA 19-9 in the presence of elevated SDC1. DISCUSSION These findings suggest serum SDC1 as a promising novel biomarker for early blood-based diagnosis of pancreatic cancer.
Collapse
Affiliation(s)
- Doron Yablecovitch
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Shomron Ben-Horin
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Orit Picard
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Miri Yavzori
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Ella Fudim
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Moshe Nadler
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Idan Levy
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Emad Sakhnini
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Alon Lang
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Tal Engel
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Maor Lahav
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Talia Saker
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
- Shalvata Mental Health Center, Hod Hasharon, Israel;
| | - Sandra Neuman
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Limor Selinger
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Revital Dvir
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| | - Maria Raitses-Gurevich
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
- Department of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel.
| | - Talia Golan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
- Department of Oncology, Chaim Sheba Medical Center, Tel Hashomer, Israel.
| | - Ido Laish
- Gastroenterology Institute, Chaim Sheba Medical Center, Tel Hashomer, Israel;
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel;
| |
Collapse
|
20
|
Santos NJ, Barquilha CN, Barbosa IC, Macedo RT, Lima FO, Justulin LA, Barbosa GO, Carvalho HF, Felisbino SL. Syndecan Family Gene and Protein Expression and Their Prognostic Values for Prostate Cancer. Int J Mol Sci 2021; 22:ijms22168669. [PMID: 34445387 PMCID: PMC8395474 DOI: 10.3390/ijms22168669] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the leading cause of cancer-associated mortality in men, and new biomarkers are still needed. The expression pattern and protein tissue localization of proteoglycans of the syndecan family (SDC 1-4) and syntenin-1 (SDCBP) were determined in normal and prostatic tumor tissue from two genetically engineered mouse models and human prostate tumors. Studies were validated using SDC 1-4 and SDCBP mRNA levels and patient survival data from The Cancer Genome Atlas and CamCAP databases. RNAseq showed increased expression of Sdc1 in Pb-Cre4/Ptenf/f mouse Pca and upregulation of Sdc3 expression and downregulation of Sdc2 and Sdc4 when compared to the normal prostatic tissue in Pb-Cre4/Trp53f/f-;Rb1f/f mouse tumors. These changes were confirmed by immunohistochemistry. In human PCa, SDC 1-4 and SDCBP immunostaining showed variable localization. Furthermore, Kaplan-Meier analysis showed that patients expressing SDC3 had shorter prostate-specific survival than those without SDC3 expression (log-rank test, p = 0.0047). Analysis of the MSKCC-derived expression showed that SDC1 and SDC3 overexpression is predictive of decreased biochemical recurrence-free survival (p = 0.0099 and p = 0.045, respectively), and SDC4 overexpression is predictive of increased biochemical recurrence-free survival (p = 0.035). SDC4 overexpression was associated with a better prognosis, while SDC1 and SDC3 were associated with more aggressive tumors and a worse prognosis.
Collapse
Affiliation(s)
- Nilton José Santos
- Department of Structural and Functional BIology, Institute of Bioscience of Botucatu (IBB), São Paulo State University, Botucatu 18618-689, SP, Brazil; (N.J.S.); (C.N.B.); (I.C.B.); (L.A.J.)
- Department of Structural and Functional Biology, Institute of Biology (IB), UNICAMP—State University of Campinas, Campinas 13083-970, SP, Brazil; (G.O.B.); (H.F.C.)
| | - Caroline Nascimento Barquilha
- Department of Structural and Functional BIology, Institute of Bioscience of Botucatu (IBB), São Paulo State University, Botucatu 18618-689, SP, Brazil; (N.J.S.); (C.N.B.); (I.C.B.); (L.A.J.)
- Department of Structural and Functional Biology, Institute of Biology (IB), UNICAMP—State University of Campinas, Campinas 13083-970, SP, Brazil; (G.O.B.); (H.F.C.)
| | - Isabela Correa Barbosa
- Department of Structural and Functional BIology, Institute of Bioscience of Botucatu (IBB), São Paulo State University, Botucatu 18618-689, SP, Brazil; (N.J.S.); (C.N.B.); (I.C.B.); (L.A.J.)
- Department of Structural and Functional Biology, Institute of Biology (IB), UNICAMP—State University of Campinas, Campinas 13083-970, SP, Brazil; (G.O.B.); (H.F.C.)
| | - Rodrigo Tavares Macedo
- Botucatu School of Medicine (FMB), São Paulo State University, Botucatu 01049-010, SP, Brazil; (R.T.M.); (F.O.L.)
| | - Flávio Oliveira Lima
- Botucatu School of Medicine (FMB), São Paulo State University, Botucatu 01049-010, SP, Brazil; (R.T.M.); (F.O.L.)
| | - Luis Antônio Justulin
- Department of Structural and Functional BIology, Institute of Bioscience of Botucatu (IBB), São Paulo State University, Botucatu 18618-689, SP, Brazil; (N.J.S.); (C.N.B.); (I.C.B.); (L.A.J.)
| | - Guilherme Oliveira Barbosa
- Department of Structural and Functional Biology, Institute of Biology (IB), UNICAMP—State University of Campinas, Campinas 13083-970, SP, Brazil; (G.O.B.); (H.F.C.)
| | - Hernandes F. Carvalho
- Department of Structural and Functional Biology, Institute of Biology (IB), UNICAMP—State University of Campinas, Campinas 13083-970, SP, Brazil; (G.O.B.); (H.F.C.)
| | - Sérgio Luis Felisbino
- Department of Structural and Functional BIology, Institute of Bioscience of Botucatu (IBB), São Paulo State University, Botucatu 18618-689, SP, Brazil; (N.J.S.); (C.N.B.); (I.C.B.); (L.A.J.)
- Correspondence:
| |
Collapse
|
21
|
Syndecan-1 (CD138), Carcinomas and EMT. Int J Mol Sci 2021; 22:ijms22084227. [PMID: 33921767 PMCID: PMC8072910 DOI: 10.3390/ijms22084227] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Cell surface proteoglycans are known to be important regulators of many aspects of cell behavior. The principal family of transmembrane proteoglycans is the syndecans, of which there are four in mammals. Syndecan-1 is mostly restricted to epithelia, and bears heparan sulfate chains that are capable of interacting with a large array of polypeptides, including extracellular matrix components and potent mediators of proliferation, adhesion and migration. For this reason, it has been studied extensively with respect to carcinomas and tumor progression. Frequently, but not always, syndecan-1 levels decrease as tumor grade, stage and invasiveness and dedifferentiation increase. This parallels experiments that show depletion of syndecan-1 can be accompanied by loss of cadherin-mediated adhesion. However, in some tumors, levels of syndecan-1 increase, but the characterization of its distribution is relevant. There can be loss of membrane staining, but acquisition of cytoplasmic and/or nuclear staining that is abnormal. Moreover, the appearance of syndecan-1 in the tumor stroma, either associated with its cellular component or the collagenous matrix, is nearly always a sign of poor prognosis. Given its relevance to myeloma progression, syndecan-1-directed antibody—toxin conjugates are being tested in clinical and preclinical trials, and may have future relevance to some carcinomas.
Collapse
|