1
|
Zhao KY, Du YX, Cao HM, Su LY, Su XL, Li X. The biological macromolecules constructed Matrigel for cultured organoids in biomedical and tissue engineering. Colloids Surf B Biointerfaces 2025; 247:114435. [PMID: 39647422 DOI: 10.1016/j.colsurfb.2024.114435] [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: 10/08/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
Matrigel is the most commonly used matrix for 3D organoid cultures. Research on the biomaterial basis of Matrigel for organoid cultures is a highly challenging field. Currently, many studies focus on Matrigel-based biological macromolecules or combinations to construct natural Matrigel and synthetic hydrogel scaffolds based on collagen, peptides, polysaccharides, microbial transglutaminase, DNA supramolecules, and polymers for organoid culture. In this review, we discuss the limitations of both natural and synthetic Matrigel, and describe alternative scaffolds that have been employed for organoid cultures. The patient-derived organoids were constructed in different cancer types and limitations of animal-derived organoids based on the hydrogel or Matrigel. The constructed techniques utilizing 3D bioprinting platforms, air-liquid interface (ALI) culture, microfluidic culture, and organ-on-a-chip platform are summarized. Given the potential of organoids for a wide range of therapeutic, tissue engineering and pharmaceutical applications, it is indeed imperative to develop defined and customized hydrogels in addition to Matrigel.
Collapse
Affiliation(s)
- Ke-Yu Zhao
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China
| | - Yi-Xiang Du
- Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Hui-Min Cao
- Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Li-Ya Su
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China
| | - Xiu-Lan Su
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China
| | - Xian Li
- Key Laboratory of Medical Cell Biology in Inner Mongolia, Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, China; Key Laboratory of Medical Cell Biology in Inner Mongolia, Inner Mongolia Bioactive Peptide Engineering Laboratory, 1 North Tongdao Street, Hohhot, Inner Mongolia 010050, China.
| |
Collapse
|
2
|
Diab L, Al Kattar S, Oueini N, Hawi J, Chrabieh A, Dosh L, Jurjus R, Leone A, Jurjus A. Syndecan-1: a key player in health and disease. Immunogenetics 2024; 77:9. [PMID: 39688651 DOI: 10.1007/s00251-024-01366-4] [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: 11/04/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024]
Abstract
Syndecan-1 (SDC-1) is a transmembrane protein localized on the basolateral surface of epithelial cells, encompassing a core protein with heparin sulfate and chondroitin sulfate glycosaminoglycan side chains. SDC-1 is involved in a panoply of cellular mechanisms including cell-to-cell adhesion, extracellular matrix interactions, cell cycle modulation, and lipid clearance. Alterations in the expression and function of SDC-1 are implicated in numerous disease entities, making it an attractive diagnostic and therapeutic target. However, despite its broad involvement in several disease processes, the underlying mechanism contributing to its diverse functions, pathogenesis, and therapeutic uses remains underexplored. Therefore, this review examines the role of SDC-1 in health and disease, focusing on liver pathologies, inflammatory diseases, infectious diseases, and cancer, and sheds light on SDC-1-based therapeutic approaches. Moreover, it delves into the mechanisms through which SDC-1 contributes to these diseases, emphasizing cell-type specific mechanisms. By comprehensively summarizing the significance of SDC-1, its association with several diseases, and its underlying mechanisms of action, the findings of this review could inform future research directions toward the development of targeted therapies and early diagnosis for a multitude of disease entities.
Collapse
Affiliation(s)
- Lara Diab
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Sahar Al Kattar
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Naim Oueini
- Department of Agriculture and Food Engineering, School of Engineering, Holy Spirit University, Kaslik, Jounieh, Lebanon
| | - Jihad Hawi
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Antoine Chrabieh
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Laura Dosh
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Rosalyn Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon
| | - Angelo Leone
- Department of Biomedicine, Neuroscience and Advanced Diagnostic, University of Palermo, Palermo, Italy
| | - Abdo Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
3
|
Peters K, Staehlke S, Rebl H, Jonitz-Heincke A, Hahn O. Impact of Metal Ions on Cellular Functions: A Focus on Mesenchymal Stem/Stromal Cell Differentiation. Int J Mol Sci 2024; 25:10127. [PMID: 39337612 PMCID: PMC11432215 DOI: 10.3390/ijms251810127] [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: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Metals play a crucial role in the human body, especially as ions in metalloproteins. Essential metals, such as calcium, iron, and zinc are crucial for various physiological functions, but their interactions within biological networks are complex and not fully understood. Mesenchymal stem/stromal cells (MSCs) are essential for tissue regeneration due to their ability to differentiate into various cell types. This review article addresses the effects of physiological and unphysiological, but not directly toxic, metal ion concentrations, particularly concerning MSCs. Overloading or unbalancing of metal ion concentrations can significantly impair the function and differentiation capacity of MSCs. In addition, excessive or unbalanced metal ion concentrations can lead to oxidative stress, which can affect viability or inflammation. Data on the effects of metal ions on MSC differentiation are limited and often contradictory. Future research should, therefore, aim to clarify the mechanisms by which metal ions affect MSC differentiation, focusing on aspects such as metal ion interactions, ion concentrations, exposure duration, and other environmental conditions. Understanding these interactions could ultimately improve the design of biomaterials and implants to promote MSC-mediated tissue regeneration. It could also lead to the development of innovative therapeutic strategies in regenerative medicine.
Collapse
Affiliation(s)
- Kirsten Peters
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Susanne Staehlke
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Henrike Rebl
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| | - Anika Jonitz-Heincke
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopaedics, Rostock University Medical Center, Doberaner Strasse 142, 18057 Rostock, Germany;
| | - Olga Hahn
- Institute of Cell Biology, Rostock University Medical Center Rostock, Schillingallee 69, 18057 Rostock, Germany; (S.S.); (H.R.); (O.H.)
| |
Collapse
|
4
|
Ricard-Blum S, Vivès RR, Schaefer L, Götte M, Merline R, Passi A, Heldin P, Magalhães A, Reis CA, Skandalis SS, Karamanos NK, Perez S, Nikitovic D. A biological guide to glycosaminoglycans: current perspectives and pending questions. FEBS J 2024; 291:3331-3366. [PMID: 38500384 DOI: 10.1111/febs.17107] [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: 10/10/2023] [Revised: 01/08/2024] [Accepted: 02/20/2024] [Indexed: 03/20/2024]
Abstract
Mammalian glycosaminoglycans (GAGs), except hyaluronan (HA), are sulfated polysaccharides that are covalently attached to core proteins to form proteoglycans (PGs). This article summarizes key biological findings for the most widespread GAGs, namely HA, chondroitin sulfate/dermatan sulfate (CS/DS), keratan sulfate (KS), and heparan sulfate (HS). It focuses on the major processes that remain to be deciphered to get a comprehensive view of the mechanisms mediating GAG biological functions. They include the regulation of GAG biosynthesis and postsynthetic modifications in heparin (HP) and HS, the composition, heterogeneity, and function of the tetrasaccharide linkage region and its role in disease, the functional characterization of the new PGs recently identified by glycoproteomics, the selectivity of interactions mediated by GAG chains, the display of GAG chains and PGs at the cell surface and their impact on the availability and activity of soluble ligands, and on their move through the glycocalyx layer to reach their receptors, the human GAG profile in health and disease, the roles of GAGs and particular PGs (syndecans, decorin, and biglycan) involved in cancer, inflammation, and fibrosis, the possible use of GAGs and PGs as disease biomarkers, and the design of inhibitors targeting GAG biosynthetic enzymes and GAG-protein interactions to develop novel therapeutic approaches.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- Univ Lyon 1, ICBMS, UMR 5246 University Lyon 1 - CNRS, Villeurbanne cedex, France
| | | | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Germany
| | - Rosetta Merline
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | | | - Paraskevi Heldin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Serge Perez
- Centre de Recherche sur les Macromolécules Végétales, University of Grenoble-Alpes, CNRS, France
| | - Dragana Nikitovic
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
5
|
Balistreri CR, Di Giorgi L, Monastero R. Focus of endothelial glycocalyx dysfunction in ischemic stroke and Alzheimer's disease: Possible intervention strategies. Ageing Res Rev 2024; 99:102362. [PMID: 38830545 DOI: 10.1016/j.arr.2024.102362] [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: 04/22/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
The integrity of the endothelial glycocalyx (eGCX), a mixture of carbohydrates attached to proteins expressed on the surface of blood vessel endothelial cells (EC), is critical for the maintenance of homeostasis of the cardiovascular system and all systems of the human body, the endothelium being the critical component of the stroma of all tissues. Consequently, dysfunction of eGCX results in a dysfunctional cardiovascular wall and severe downstream cardiovascular events, which contribute to the onset of cardio- and cerebrovascular diseases and neurodegenerative disorders, as well as other age-related diseases (ARDs). The key role of eGCX dysfunction in the onset of ARDs is examined here, with a focus on the most prevalent neurological diseases: ischemic stroke and Alzheimer's disease. Furthermore, the advantages and limitations of some treatment strategies for anti-eGCX dysfunction are described, ranging from experimental drug therapies, which need to be better tested and explored not only in animal models but also in humans, as well as reprogramming, the use of nutraceuticals, which are emerging as regenerative and new approaches. The promotion of these strategies is essential to keep eGCX and endothelium healthy, as is the development of intravital (e.g., intravascular) tools to estimate eGCX health status and treatment efficacy, which could lead to advanced solutions to address ARDs.
Collapse
Affiliation(s)
- Carmela Rita Balistreri
- Cellular, Molecular and Clinical Pathological Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Palermo 90134, Italy.
| | - Lucia Di Giorgi
- Memory and Parkinson's disease Center Policlinico "Paolo Giaccone", Palermo, and Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Via La Loggia 1, Palermo 90129, Italy
| | - Roberto Monastero
- Memory and Parkinson's disease Center Policlinico "Paolo Giaccone", Palermo, and Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, Via La Loggia 1, Palermo 90129, Italy.
| |
Collapse
|
6
|
Su C, Mo J, Dong S, Liao Z, Zhang B, Zhu P. Integrinβ-1 in disorders and cancers: molecular mechanisms and therapeutic targets. Cell Commun Signal 2024; 22:71. [PMID: 38279122 PMCID: PMC10811905 DOI: 10.1186/s12964-023-01338-3] [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: 08/23/2023] [Accepted: 09/27/2023] [Indexed: 01/28/2024] Open
Abstract
Integrinβ-1 (ITGB1) is a crucial member of the transmembrane glycoprotein signaling receptor family and is also central to the integrin family. It forms heterodimers with other ligands, participates in intracellular signaling and controls a variety of cellular processes, such as angiogenesis and the growth of neurons; because of its role in bidirectional signaling regulation both inside and outside the membrane, ITGB1 must interact with a multitude of substances, so a variety of interfering factors can affect ITGB1 and lead to changes in its function. Over the past 20 years, many studies have confirmed a clear causal relationship between ITGB1 dysregulation and cancer development and progression in a wide range of benign diseases and solid tumor types, which may imply that ITGB1 is a prognostic biomarker and a therapeutic target for cancer treatment that warrants further investigation. This review summarizes the biological roles of ITGB1 in benign diseases and cancers, and compiles the current status of ITGB1 function and therapy in various aspects of tumorigenesis and progression. Finally, future research directions and application prospects of ITGB1 are suggested. Video Abstract.
Collapse
Affiliation(s)
- Chen Su
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Jie Mo
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Shuilin Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China
| | - Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, People's Republic of China.
| | - Peng Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, Hubei, People's Republic of China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
7
|
Velho RV, Danielyan I, Mechsner S, Götte M. Sox-2 positive cells identified in lymph nodes from endometriosis patients may play a role in the disease pathogenesis. Eur J Obstet Gynecol Reprod Biol 2023; 288:124-129. [PMID: 37506598 DOI: 10.1016/j.ejogrb.2023.07.017] [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: 06/09/2023] [Revised: 07/18/2023] [Accepted: 07/24/2023] [Indexed: 07/30/2023]
Abstract
OBJECTIVE This study aimed to characterize Sox-2 in sentinel lymph nodes and randomly obtained lymph nodes from endometriosis (EM) patients for the first time. STUDY DESIGN This prospective study analyzed tissue samples from surgical specimens collected from May until December 2007 in the Endometriosis Center Charité, Berlin. Lymph node samples from 38 women aged between 22 and 49 years who underwent laparoscopy due to symptomatic EM were analyzed. The material was obtained either randomly or, in the case of deep infiltrating endometriosis, detected using 4 cc Patent Blue®, labeled intraoperatively, which made the sentinel lymph nodes available for histological examination. Together with hematoxylin and eosin staining, the sections were evaluated by immunohistochemistry with antibodies against estrogen and progesterone receptors and Sox-2. Using double-immunofluorescence microscopy, the colocalization of Sox-2 and estrogen receptors were evaluated. RESULTS Sox-2-positive cells were identified in the lymph nodes' cortical and medullary zones, with a higher expression in the medullary layer. Occasionally, Sox-2 positive stained cell groups, called cell nests, could also be detected. The number of Sox-2 positive cells in the sentinel lymph nodes was almost three times higher than in the random lymph nodes (p = 0.031). A significant five-fold increase (p = 0.0013) in Sox-2 expression was seen in the estrogen and progesterone receptor (ER/PR) positive patient group compared to the progesterone receptor positive group or hormone receptor negative patients. Identical hormone-related Sox-2 expression was also detected separately for the sentinel lymph node group (p = 0.0174). Sox-2 showed pronounced colocalisation with estrogen receptors. CONCLUSION The lymphatic involvement in EM is evidence of a systemic disease manifestation and provides evidence of an immune system failure. In recent years, many theories have been studied, but there is no single theory that could explain all aspects of EM. The future concept of EM is likely to incorporate the elements from all the pathogenetic theories already described. Through this study, stem cells and lymphatic metastasis theories were incorporated.
Collapse
Affiliation(s)
- Renata Voltolini Velho
- Department of Gynecology Charité with Center of Oncological Surgery, Endometriosis Research Center Charité, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Inna Danielyan
- Department of Gynecology and Obstetrics, Münster University Hospital, Labor PAN-Zentrum, Vesaliusweg 2-4, 48149 Münster, Germany
| | - Sylvia Mechsner
- Department of Gynecology Charité with Center of Oncological Surgery, Endometriosis Research Center Charité, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Labor PAN-Zentrum, Vesaliusweg 2-4, 48149 Münster, Germany.
| |
Collapse
|
8
|
Benjamin M, Malakar P, Sinha RA, Nasser MW, Batra SK, Siddiqui JA, Chakravarti B. Molecular signaling network and therapeutic developments in breast cancer brain metastasis. ADVANCES IN CANCER BIOLOGY - METASTASIS 2023; 7:100079. [PMID: 36536947 PMCID: PMC7613958 DOI: 10.1016/j.adcanc.2022.100079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Breast cancer (BC) is one of the most frequently diagnosed cancers in women worldwide. It has surpassed lung cancer as the leading cause of cancer-related death. Breast cancer brain metastasis (BCBM) is becoming a major clinical concern that is commonly associated with ER-ve and HER2+ve subtypes of BC patients. Metastatic lesions in the brain originate when the cancer cells detach from a primary breast tumor and establish metastatic lesions and infiltrate near and distant organs via systemic blood circulation by traversing the BBB. The colonization of BC cells in the brain involves a complex interplay in the tumor microenvironment (TME), metastatic cells, and brain cells like endothelial cells, microglia, and astrocytes. BCBM is a significant cause of morbidity and mortality and presents a challenge to developing successful cancer therapy. In this review, we discuss the molecular mechanism of BCBM and novel therapeutic strategies for patients with brain metastatic BC.
Collapse
Affiliation(s)
- Mercilena Benjamin
- Lab Oncology, Dr. B.R.A.I.R.C.H. All India Institute of Medical Sciences, New Delhi, India
| | - Pushkar Malakar
- Department of Biomedical Science and Technology, School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute, Narendrapur, West Bengal, 700103, India
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Jawed Akhtar Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68108, USA
| | - Bandana Chakravarti
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India
| |
Collapse
|
9
|
Yang H, Wang L. Heparan sulfate proteoglycans in cancer: Pathogenesis and therapeutic potential. Adv Cancer Res 2023; 157:251-291. [PMID: 36725112 DOI: 10.1016/bs.acr.2022.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The heparan sulfate proteoglycans (HSPGs) are glycoproteins that consist of a proteoglycan "core" protein and covalently attached heparan sulfate (HS) chain. HSPGs are ubiquitously expressed in mammalian cells on the cell surface and in the extracellular matrix (ECM) and secretory vesicles. Within HSPGs, the protein cores determine when and where HSPG expression takes place, and the HS chains mediate most of HSPG's biological roles through binding various protein ligands, including cytokines, chemokines, growth factors and receptors, morphogens, proteases, protease inhibitors, and ECM proteins. Through these interactions, HSPGs modulate cell proliferation, adhesion, migration, invasion, and angiogenesis to display essential functions in physiology and pathology. Under physiological conditions, the expression and localization of HSPGs are finely regulated to orchestrate their physiological functions, and this is disrupted in cancer. The HSPG dysregulation elicits multiple oncogenic signaling, including growth factor signaling, ECM and Integrin signaling, chemokine and immune signaling, cancer stem cell, cell differentiation, apoptosis, and senescence, to prompt cell transformation, proliferation, tumor invasion and metastasis, tumor angiogenesis and inflammation, and immunotolerance. These oncogenic roles make HSPGs an attractive pharmacological target for anti-cancer therapy. Several therapeutic strategies have been under development, including anti-HSPG antibodies, peptides and HS mimetics, synthetic xylosides, and heparinase inhibitors, and shown promising anti-cancer efficacy. Therefore, much progress has been made in this line of study. However, it needs to bear in mind that the roles of HSPGs in cancer can be either oncogenic or tumor-suppressive, depending on the HSPG and the cancer cell type with the underlying mechanisms that remain obscure. Further studies need to address these to fill the knowledge gap and rationalize more efficient therapeutic targeting.
Collapse
Affiliation(s)
- Hua Yang
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Lianchun Wang
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; Bryd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.
| |
Collapse
|
10
|
Ibrahim SA, Yip GW, Götte M. Editorial: Cancer stem cells as attractive targets for breast cancer therapy. Front Oncol 2023; 13:1151742. [PMID: 36937444 PMCID: PMC10020607 DOI: 10.3389/fonc.2023.1151742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Affiliation(s)
- Sherif Abdelaziz Ibrahim
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
- *Correspondence: Sherif Abdelaziz Ibrahim, ; ; Martin Götte,
| | - George W. Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
- *Correspondence: Sherif Abdelaziz Ibrahim, ; ; Martin Götte,
| |
Collapse
|
11
|
Hua SH, Viera M, Yip GW, Bay BH. Theranostic Applications of Glycosaminoglycans in Metastatic Renal Cell Carcinoma. Cancers (Basel) 2022; 15:cancers15010266. [PMID: 36612261 PMCID: PMC9818616 DOI: 10.3390/cancers15010266] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Renal cell carcinoma (RCC) makes up the majority of kidney cancers, with a poor prognosis for metastatic RCC (mRCC). Challenges faced in the management of mRCC, include a lack of reliable prognostic markers and biomarkers for precise monitoring of disease treatment, together with the potential risk of toxicity associated with more recent therapeutic options. Glycosaminoglycans (GAGs) are a class of carbohydrates that can be categorized into four main subclasses, viz., chondroitin sulfate, hyaluronic acid, heparan sulfate and keratan sulfate. GAGs are known to be closely associated with cancer progression and modulation of metastasis by modification of the tumor microenvironment. Alterations of expression, composition and spatiotemporal distribution of GAGs in the extracellular matrix (ECM), dysregulate ECM functions and drive cancer invasion. In this review, we focus on the clinical utility of GAGs as biomarkers for mRCC (which is important for risk stratification and strategizing effective treatment protocols), as well as potential therapeutic targets that could benefit patients afflicted with advanced RCC. Besides GAG-targeted therapies that holds promise in mRCC, other potential strategies include utilizing GAGs as drug carriers and their mimetics to counter cancer progression, and enhance immunotherapy through binding and transducing signals for immune mediators.
Collapse
|
12
|
Liu Y, Wang Y, Sun S, Chen Z, Xiang S, Ding Z, Huang Z, Zhang B. Understanding the versatile roles and applications of EpCAM in cancers: from bench to bedside. Exp Hematol Oncol 2022; 11:97. [PMID: 36369033 PMCID: PMC9650829 DOI: 10.1186/s40164-022-00352-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial cell adhesion molecule (EpCAM) functions not only in physiological processes but also participates in the development and progression of cancer. In recent decades, extensive efforts have been made to decipher the role of EpCAM in cancers. Great advances have been achieved in elucidating its structure, molecular functions, pathophysiological mechanisms, and clinical applications. Beyond its well-recognized role as a biomarker of cancer stem cells (CSCs) or circulating tumor cells (CTCs), EpCAM exhibits novel and promising value in targeted therapy. At the same time, the roles of EpCAM in cancer progression are found to be highly context-dependent and even contradictory in some cases. The versatile functional modules of EpCAM and its communication with other signaling pathways complicate the study of this molecule. In this review, we start from the structure of EpCAM and focus on communication with other signaling pathways. The impacts on the biology of cancers and the up-to-date clinical applications of EpCAM are also introduced and summarized, aiming to shed light on the translational prospects of EpCAM.
Collapse
Affiliation(s)
- Yiyang Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufei Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Sun
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeyu Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Xiang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeyang Ding
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhao Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, National Health Commission, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
13
|
Zhang Z, Li J, Jiao S, Han G, Zhu J, Liu T. Functional and clinical characteristics of focal adhesion kinases in cancer progression. Front Cell Dev Biol 2022; 10:1040311. [PMID: 36407100 PMCID: PMC9666724 DOI: 10.3389/fcell.2022.1040311] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022] Open
Abstract
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase and an adaptor protein that primarily regulates adhesion signaling and cell migration. FAK promotes cell survival in response to stress. Increasing evidence has shown that at the pathological level, FAK is highly expressed in multiple tumors in several systems (including lung, liver, gastric, and colorectal cancers) and correlates with tumor aggressiveness and patient prognosis. At the molecular level, FAK promotes tumor progression mainly by altering survival signals, invasive capacity, epithelial-mesenchymal transition, the tumor microenvironment, the Warburg effect, and stemness of tumor cells. Many effective drugs have been developed based on the comprehensive role of FAK in tumor cells. In addition, its potential as a tumor marker cannot be ignored. Here, we discuss the pathological and pre-clinical evidence of the role of FAK in cancer development; we hope that these findings will assist in FAK-based clinical studies.
Collapse
Affiliation(s)
- Zhaoyu Zhang
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jinlong Li
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Simin Jiao
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Guangda Han
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jiaming Zhu
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tianzhou Liu
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
14
|
Sparić R, Andjić M, Babović I, Nejković L, Mitrović M, Štulić J, Pupovac M, Tinelli A. Molecular Insights in Uterine Leiomyosarcoma: A Systematic Review. Int J Mol Sci 2022; 23:ijms23179728. [PMID: 36077127 PMCID: PMC9456512 DOI: 10.3390/ijms23179728] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/28/2022] Open
Abstract
Uterine fibroids (UFs) are the most common benign tumors of female genital diseases, unlike uterine leiomyosarcoma (LMS), a rare and aggressive uterine cancer. This narrative review aims to discuss the biology and diagnosis of LMS and, at the same time, their differential diagnosis, in order to distinguish the biological and molecular origins. The authors performed a Medline and PubMed search for the years 1990–2022 using a combination of keywords on the topics to highlight the many genes and proteins involved in the pathogenesis of LMS. The mutation of these genes, in addition to the altered expression and functions of their enzymes, are potentially biomarkers of uterine LMS. Thus, the use of this molecular and protein information could favor differential diagnosis and personalized therapy based on the molecular characteristics of LMS tissue, leading to timely diagnoses and potential better outcomes for patients.
Collapse
Affiliation(s)
- Radmila Sparić
- Clinic for Gynecology and Obstetrics, University Clinical Centre of Serbia, Koste Todorovića 26, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Mladen Andjić
- Clinic for Gynecology and Obstetrics, University Clinical Centre of Serbia, Koste Todorovića 26, 11000 Belgrade, Serbia
- Correspondence: (M.A.); (A.T.)
| | - Ivana Babović
- Clinic for Gynecology and Obstetrics, University Clinical Centre of Serbia, Koste Todorovića 26, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Lazar Nejković
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Clinic of Gynecology and Obstetrics Narodni Front, 11000 Belgrade, Serbia
| | - Milena Mitrović
- Clinic for Gynecology and Obstetrics, University Clinical Centre of Serbia, Koste Todorovića 26, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Jelena Štulić
- Clinic of Gynecology and Obstetrics Narodni Front, 11000 Belgrade, Serbia
| | - Miljan Pupovac
- Clinic for Gynecology and Obstetrics, University Clinical Centre of Serbia, Koste Todorovića 26, 11000 Belgrade, Serbia
| | - Andrea Tinelli
- Department of Obstetrics and Gynecology, and CERICSAL (CEntro di Ricerca Clinico SALentino), “Verisdelli Ponti Hospital”, Via Giuseppina Delli Ponti, 73020 Scorrano, LE, Italy
- Correspondence: (M.A.); (A.T.)
| |
Collapse
|
15
|
The Cell Surface Heparan Sulfate Proteoglycan Syndecan-3 Promotes Ovarian Cancer Pathogenesis. Int J Mol Sci 2022; 23:ijms23105793. [PMID: 35628603 PMCID: PMC9145288 DOI: 10.3390/ijms23105793] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 11/24/2022] Open
Abstract
Syndecans are transmembrane heparan sulfate proteoglycans that integrate signaling at the cell surface. By interacting with cytokines, signaling receptors, proteases, and extracellular matrix proteins, syndecans regulate cell proliferation, metastasis, angiogenesis, and inflammation. We analyzed public gene expression datasets to evaluate the dysregulation and potential prognostic impact of Syndecan-3 in ovarian cancer. Moreover, we performed functional in vitro analysis in syndecan-3-siRNA-treated SKOV3 and CAOV3 ovarian cancer cells. In silico analysis of public gene array datasets revealed that syndecan-3 mRNA expression was significantly increased 5.8-fold in ovarian cancer tissues (n = 744) and 3.4-fold in metastases (n = 44) compared with control tissue (n = 46), as independently confirmed in an RNAseq dataset on ovarian serous cystadenocarcinoma tissue (n = 374, controls: n = 133, 3.5-fold increase tumor vs. normal). Syndecan-3 siRNA knockdown impaired 3D spheroid growth and colony formation as stemness-related readouts in SKOV3 and CAOV3 cells. In SKOV3, but not in CAOV3 cells, syndecan-3 depletion reduced cell viability both under basal conditions and under chemotherapy with cisplatin, or cisplatin and paclitaxel. While analysis of the SIOVDB database did not reveal differences in Syndecan-3 expression between patients, sensitive, resistant or refractory to chemotherapy, KM Plotter analysis of 1435 ovarian cancer patients revealed that high syndecan-3 expression was associated with reduced survival in patients treated with taxol and platin. At the molecular level, a reduction in Stat3 activation and changes in the expression of Wnt and notch signaling constituents were observed. Our study suggests that up-regulation of syndecan-3 promotes the pathogenesis of ovarian cancer by modulating stemness-associated pathways.
Collapse
|
16
|
Tissue extracellular matrix hydrogels as alternatives to Matrigel for culturing gastrointestinal organoids. Nat Commun 2022; 13:1692. [PMID: 35354790 PMCID: PMC8967832 DOI: 10.1038/s41467-022-29279-4] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/04/2022] [Indexed: 12/16/2022] Open
Abstract
Matrigel, a mouse tumor extracellular matrix protein mixture, is an indispensable component of most organoid tissue culture. However, it has limited the utility of organoids for drug development and regenerative medicine due to its tumor-derived origin, batch-to-batch variation, high cost, and safety issues. Here, we demonstrate that gastrointestinal tissue-derived extracellular matrix hydrogels are suitable substitutes for Matrigel in gastrointestinal organoid culture. We found that the development and function of gastric or intestinal organoids grown in tissue extracellular matrix hydrogels are comparable or often superior to those in Matrigel. In addition, gastrointestinal extracellular matrix hydrogels enabled long-term subculture and transplantation of organoids by providing gastrointestinal tissue-mimetic microenvironments. Tissue-specific and age-related extracellular matrix profiles that affect organoid development were also elucidated through proteomic analysis. Together, our results suggest that extracellular matrix hydrogels derived from decellularized gastrointestinal tissues are effective alternatives to the current gold standard, Matrigel, and produce organoids suitable for gastrointestinal disease modeling, drug development, and tissue regeneration.
Collapse
|
17
|
Hilgers K, Ibrahim SA, Kiesel L, Greve B, Espinoza-Sánchez NA, Götte M. Differential Impact of Membrane-Bound and Soluble Forms of the Prognostic Marker Syndecan-1 on the Invasiveness, Migration, Apoptosis, and Proliferation of Cervical Cancer Cells. Front Oncol 2022; 12:803899. [PMID: 35155241 PMCID: PMC8828476 DOI: 10.3389/fonc.2022.803899] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/03/2022] [Indexed: 12/16/2022] Open
Abstract
Cervical cancer ranks fourth among the most commonly diagnosed malignant tumors in women worldwide. Previously published evidence suggested a possible connection between the expression of the membrane-bound heparan sulfate proteoglycan syndecan-1 (Sdc-1) and the development of cervical carcinoma. Sdc-1 serves as a matrix receptor and coreceptor for receptor tyrosine kinases and additional signaling pathways. It influences cell proliferation, adhesion, and migration and is seen as a modulator of the tumor microenvironment. Following proteolytic cleavage of its extracellular domain in a process called shedding, Sdc-1 can act as a paracrine effector. The loss of Sdc-1 expression is associated with low differentiation of cervical carcinoma and with an increased rate of lymph node metastases. Here, we analyzed the clinical impact of Sdc-1 expression by analysis of public gene expression datasets and studied the effect of an overexpression of Sdc-1 and its membrane-bound and soluble forms on the malignant properties of the human cervical carcinoma cell line HeLa through functional analysis. For this purpose, the HeLa cells were stably transfected with the control plasmid pcDNA3.1 and three different Sdc-1-DNA constructs,encoding wild-type, permanently membrane-bound, and constitutively soluble Sdc-1. In clinical specimens, Sdc-1 mRNA was more highly expressed in local tumor tissues than in normal and metastatic cervical cancer tissues. Moreover, high Sdc-1 expression correlated with a poor prognosis in Kaplan-Meier survival analysis, suggesting the important role of Sdc-1 in the progression of this type of cancer. In vitro, we found that the soluble, as well as the permanently membrane-bound forms of Sdc-1 modulated the proliferation and the cell cycle, while membrane-bound Sdc1 regulated HeLa cell apoptosis. The overexpression of Sdc-1 and its soluble form increased invasiveness. In vitro scratch/wound healing assay, showed reduced Sdc-1-dependent cell motility which was linked to the Rho-GTPase signaling pathway. In conclusion, in cervical cancer Sdc-1 modulates pathogenetically relevant processes, which depend on the membrane-association of Sdc-1.
Collapse
Affiliation(s)
- Katharina Hilgers
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | | | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - Nancy A Espinoza-Sánchez
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.,Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| |
Collapse
|
18
|
Marques-Magalhães Â, Cruz T, Costa ÂM, Estêvão D, Rios E, Canão PA, Velho S, Carneiro F, Oliveira MJ, Cardoso AP. Decellularized Colorectal Cancer Matrices as Bioactive Scaffolds for Studying Tumor-Stroma Interactions. Cancers (Basel) 2022; 14:cancers14020359. [PMID: 35053521 PMCID: PMC8773780 DOI: 10.3390/cancers14020359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/02/2022] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
More than a physical structure providing support to tissues, the extracellular matrix (ECM) is a complex and dynamic network of macromolecules that modulates the behavior of both cancer cells and associated stromal cells of the tumor microenvironment (TME). Over the last few years, several efforts have been made to develop new models that accurately mimic the interconnections within the TME and specifically the biomechanical and biomolecular complexity of the tumor ECM. Particularly in colorectal cancer, the ECM is highly remodeled and disorganized and constitutes a key component that affects cancer hallmarks, such as cell differentiation, proliferation, angiogenesis, invasion and metastasis. Therefore, several scaffolds produced from natural and/or synthetic polymers and ceramics have been used in 3D biomimetic strategies for colorectal cancer research. Nevertheless, decellularized ECM from colorectal tumors is a unique model that offers the maintenance of native ECM architecture and molecular composition. This review will focus on innovative and advanced 3D-based models of decellularized ECM as high-throughput strategies in colorectal cancer research that potentially fill some of the gaps between in vitro 2D and in vivo models. Our aim is to highlight the need for strategies that accurately mimic the TME for precision medicine and for studying the pathophysiology of the disease.
Collapse
Affiliation(s)
- Ângela Marques-Magalhães
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Tânia Cruz
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Ângela Margarida Costa
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
| | - Diogo Estêvão
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Elisabete Rios
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- IPATIMUP-Institute of Pathology and Molecular Immunology, University of Porto, 4200-135 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário São João, 4200-319 Porto, Portugal
| | - Pedro Amoroso Canão
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário São João, 4200-319 Porto, Portugal
| | - Sérgia Velho
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- IPATIMUP-Institute of Pathology and Molecular Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Fátima Carneiro
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- IPATIMUP-Institute of Pathology and Molecular Immunology, University of Porto, 4200-135 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
- Department of Pathology, Centro Hospitalar Universitário São João, 4200-319 Porto, Portugal
| | - Maria José Oliveira
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal;
| | - Ana Patrícia Cardoso
- i3S-Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (Â.M.-M.); (T.C.); (Â.M.C.); (D.E.); (E.R.); (S.V.); (F.C.); (M.J.O.)
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- Correspondence: ; Tel.: +351-22-607-4900
| |
Collapse
|
19
|
Hassanein SS, Abdel-Mawgood AL, Ibrahim SA. EGFR-Dependent Extracellular Matrix Protein Interactions Might Light a Candle in Cell Behavior of Non-Small Cell Lung Cancer. Front Oncol 2021; 11:766659. [PMID: 34976811 PMCID: PMC8714827 DOI: 10.3389/fonc.2021.766659] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022] Open
Abstract
Lung cancer remains the leading cause of cancer-related death and is associated with a poor prognosis. Lung cancer is divided into 2 main types: the major in incidence is non-small cell lung cancer (NSCLC) and the minor is small cell lung cancer (SCLC). Although NSCLC progression depends on driver mutations, it is also affected by the extracellular matrix (ECM) interactions that activate their corresponding signaling molecules in concert with integrins and matrix metalloproteinases (MMPs). These signaling molecules include cytoplasmic kinases, small GTPases, adapter proteins, and receptor tyrosine kinases (RTKs), particularly the epidermal growth factor receptor (EGFR). In NSCLC, the interplay between ECM and EGFR regulates ECM stiffness, angiogenesis, survival, adhesion, migration, and metastasis. Furthermore, some tumor-promoting ECM components (e.g., glycoproteins and proteoglycans) enhance activation of EGFR and loss of PTEN. On the other hand, other tumor-suppressing glycoproteins and -proteoglycans can inhibit EGFR activation, suppressing cell invasion and migration. Therefore, deciphering the molecular mechanisms underlying EGFR and ECM interactions might provide a better understanding of disease pathobiology and aid in developing therapeutic strategies. This review critically discusses the crosstalk between EGFR and ECM affecting cell behavior of NSCLC, as well as the involvement of ECM components in developing resistance to EGFR inhibition.
Collapse
Affiliation(s)
- Sarah Sayed Hassanein
- Biotechnology Program, Basic and Applied Sciences (BAS) Institute, Egypt-Japan University of Science and Technology (E-JUST), Alexandria, Egypt
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Ahmed Lotfy Abdel-Mawgood
- Biotechnology Program, Basic and Applied Sciences (BAS) Institute, Egypt-Japan University of Science and Technology (E-JUST), Alexandria, Egypt
| | | |
Collapse
|
20
|
Kim MS, Ha SE, Wu M, Zogg H, Ronkon CF, Lee MY, Ro S. Extracellular Matrix Biomarkers in Colorectal Cancer. Int J Mol Sci 2021; 22:ijms22179185. [PMID: 34502094 PMCID: PMC8430714 DOI: 10.3390/ijms22179185] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/12/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022] Open
Abstract
The cellular microenvironment composition and changes therein play an extremely important role in cancer development. Changes in the extracellular matrix (ECM), which constitutes a majority of the tumor stroma, significantly contribute to the development of the tumor microenvironment. These alterations within the ECM and formation of the tumor microenvironment ultimately lead to tumor development, invasion, and metastasis. The ECM is composed of various molecules such as collagen, elastin, laminin, fibronectin, and the MMPs that cleave these protein fibers and play a central role in tissue remodeling. When healthy cells undergo an insult like DNA damage and become cancerous, if the ECM does not support these neoplastic cells, further development, invasion, and metastasis fail to occur. Therefore, ECM-related cancer research is indispensable, and ECM components can be useful biomarkers as well as therapeutic targets. Colorectal cancer specifically, is also affected by the ECM and many studies have been conducted to unravel the complex association between the two. Here we summarize the importance of several ECM components in colorectal cancer as well as their potential roles as biomarkers.
Collapse
Affiliation(s)
- Min-Seob Kim
- Department of Physiology, Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Korea; (M.-S.K.); (M.W.)
| | - Se-Eun Ha
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA; (S.-E.H.); (H.Z.); (C.F.R.)
| | - Moxin Wu
- Department of Physiology, Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Korea; (M.-S.K.); (M.W.)
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang 332000, China
| | - Hannah Zogg
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA; (S.-E.H.); (H.Z.); (C.F.R.)
| | - Charles F. Ronkon
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA; (S.-E.H.); (H.Z.); (C.F.R.)
| | - Moon-Young Lee
- Department of Physiology, Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan 54538, Korea; (M.-S.K.); (M.W.)
- Correspondence: (M.-Y.L.); (S.R.)
| | - Seungil Ro
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV 89557, USA; (S.-E.H.); (H.Z.); (C.F.R.)
- Correspondence: (M.-Y.L.); (S.R.)
| |
Collapse
|
21
|
Karamanos NK, Piperigkou Z, Passi A, Götte M, Rousselle P, Vlodavsky I. Extracellular matrix-based cancer targeting. Trends Mol Med 2021; 27:1000-1013. [PMID: 34389240 DOI: 10.1016/j.molmed.2021.07.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023]
Abstract
Tumor extracellular matrix (ECM) operates in a coordinated mode with cancer and stroma cells to evoke the multistep process of metastatic potential. The remodeled tumor-associated matrix provides a point for direct or complementary therapeutic targeting. Here, we cover and critically address the importance of ECM networks and their macromolecules in cancer. We focus on the roles of key structural and functional ECM components, and their degradation enzymes and extracellular vesicles, aiming at improving our understanding of the mechanisms contributing to tumor initiation, growth, and dissemination, and discuss potential new approaches for ECM-based therapeutic targeting and diagnosis.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece; Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece.
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece; Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Patricia Rousselle
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMR 5305, CNRS-Université Lyon 1, SFR BioSciences Gerland-Lyon Sud, 7 Passage du Vercors, Lyon, France
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
22
|
Rigiracciolo DC, Cirillo F, Talia M, Muglia L, Gutkind JS, Maggiolini M, Lappano R. Focal Adhesion Kinase Fine Tunes Multifaced Signals toward Breast Cancer Progression. Cancers (Basel) 2021; 13:645. [PMID: 33562737 PMCID: PMC7915897 DOI: 10.3390/cancers13040645] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer represents the most common diagnosed malignancy and the main leading cause of tumor-related death among women worldwide. Therefore, several efforts have been made in order to identify valuable molecular biomarkers for the prognosis and prediction of therapeutic responses in breast tumor patients. In this context, emerging discoveries have indicated that focal adhesion kinase (FAK), a non-receptor tyrosine kinase, might represent a promising target involved in breast tumorigenesis. Of note, high FAK expression and activity have been tightly correlated with a poor clinical outcome and metastatic features in several tumors, including breast cancer. Recently, a role for the integrin-FAK signaling in mechanotransduction has been suggested and the function of FAK within the breast tumor microenvironment has been ascertained toward tumor angiogenesis and vascular permeability. FAK has been also involved in cancer stem cells (CSCs)-mediated initiation, maintenance and therapeutic responses of breast tumors. In addition, the potential of FAK to elicit breast tumor-promoting effects has been even associated with the capability to modulate immune responses. On the basis of these findings, several agents targeting FAK have been exploited in diverse preclinical tumor models. Here, we recapitulate the multifaceted action exerted by FAK and its prognostic significance in breast cancer. Moreover, we highlight the recent clinical evidence regarding the usefulness of FAK inhibitors in the treatment of breast tumors.
Collapse
Affiliation(s)
- Damiano Cosimo Rigiracciolo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| | - Marianna Talia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| | - Lucia Muglia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| | - Jorge Silvio Gutkind
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA;
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (F.C.); (M.T.); (L.M.); (R.L.)
| |
Collapse
|
23
|
Barkovskaya A, Buffone A, Žídek M, Weaver VM. Proteoglycans as Mediators of Cancer Tissue Mechanics. Front Cell Dev Biol 2020; 8:569377. [PMID: 33330449 PMCID: PMC7734320 DOI: 10.3389/fcell.2020.569377] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/04/2020] [Indexed: 12/16/2022] Open
Abstract
Proteoglycans are a diverse group of molecules which are characterized by a central protein backbone that is decorated with a variety of linear sulfated glycosaminoglycan side chains. Proteoglycans contribute significantly to the biochemical and mechanical properties of the interstitial extracellular matrix where they modulate cellular behavior by engaging transmembrane receptors. Proteoglycans also comprise a major component of the cellular glycocalyx to influence transmembrane receptor structure/function and mechanosignaling. Through their ability to initiate biochemical and mechanosignaling in cells, proteoglycans elicit profound effects on proliferation, adhesion and migration. Pathologies including cancer and cardiovascular disease are characterized by perturbed expression of proteoglycans where they compromise cell and tissue behavior by stiffening the extracellular matrix and increasing the bulkiness of the glycocalyx. Increasing evidence indicates that a bulky glycocalyx and proteoglycan-enriched extracellular matrix promote malignant transformation, increase cancer aggression and alter anti-tumor therapy response. In this review, we focus on the contribution of proteoglycans to mechanobiology in the context of normal and transformed tissues. We discuss the significance of proteoglycans for therapy response, and the current experimental strategies that target proteoglycans to sensitize cancer cells to treatment.
Collapse
Affiliation(s)
- Anna Barkovskaya
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Alexander Buffone
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Martin Žídek
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Valerie M. Weaver
- Center for Bioengineering & Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Bioengineering, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Department of Therapeutic Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
24
|
Ravikumar M, Smith RAA, Nurcombe V, Cool SM. Heparan Sulfate Proteoglycans: Key Mediators of Stem Cell Function. Front Cell Dev Biol 2020; 8:581213. [PMID: 33330458 PMCID: PMC7710810 DOI: 10.3389/fcell.2020.581213] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are an evolutionarily ancient subclass of glycoproteins with exquisite structural complexity. They are ubiquitously expressed across tissues and have been found to exert a multitude of effects on cell behavior and the surrounding microenvironment. Evidence has shown that heterogeneity in HSPG composition is crucial to its functions as an essential scaffolding component in the extracellular matrix as well as a vital cell surface signaling co-receptor. Here, we provide an overview of the significance of HSPGs as essential regulators of stem cell function. We discuss the various roles of HSPGs in distinct stem cell types during key physiological events, from development through to tissue homeostasis and regeneration. The contribution of aberrant HSPG production to altered stem cell properties and dysregulated cellular homeostasis characteristic of cancer is also reviewed. Finally, we consider approaches to better understand and exploit the multifaceted functions of HSPGs in influencing stem cell characteristics for cell therapy and associated culture expansion strategies.
Collapse
Affiliation(s)
- Maanasa Ravikumar
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raymond Alexander Alfred Smith
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Victor Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore, Singapore
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
25
|
Hassan N, Greve B, Espinoza-Sánchez NA, Götte M. Cell-surface heparan sulfate proteoglycans as multifunctional integrators of signaling in cancer. Cell Signal 2020; 77:109822. [PMID: 33152440 DOI: 10.1016/j.cellsig.2020.109822] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022]
Abstract
Proteoglycans (PGs) represent a large proportion of the components that constitute the extracellular matrix (ECM). They are a diverse group of glycoproteins characterized by a covalent link to a specific glycosaminoglycan type. As part of the ECM, heparan sulfate (HS)PGs participate in both physiological and pathological processes including cell recruitment during inflammation and the promotion of cell proliferation, adhesion and motility during development, angiogenesis, wound repair and tumor progression. A key function of HSPGs is their ability to modulate the expression and function of cytokines, chemokines, growth factors, morphogens, and adhesion molecules. This is due to their capacity to act as ligands or co-receptors for various signal-transducing receptors, affecting pathways such as FGF, VEGF, chemokines, integrins, Wnt, notch, IL-6/JAK-STAT3, and NF-κB. The activation of those pathways has been implicated in the induction, progression, and malignancy of a tumor. For many years, the study of signaling has allowed for designing specific drugs targeting these pathways for cancer treatment, with very positive results. Likewise, HSPGs have become the subject of cancer research and are increasingly recognized as important therapeutic targets. Although they have been studied in a variety of preclinical and experimental models, their mechanism of action in malignancy still needs to be more clearly defined. In this review, we discuss the role of cell-surface HSPGs as pleiotropic modulators of signaling in cancer and identify them as promising markers and targets for cancer treatment.
Collapse
Affiliation(s)
- Nourhan Hassan
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany; Biotechnology Program, Department of Chemistry, Faculty of Science, Cairo University, Egypt
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Albert-Schweitzer-Campus 1, A1, 48149 Münster, Germany
| | - Nancy A Espinoza-Sánchez
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany; Department of Radiotherapy-Radiooncology, Münster University Hospital, Albert-Schweitzer-Campus 1, A1, 48149 Münster, Germany.
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.
| |
Collapse
|
26
|
Teixeira FCOB, Vijaya Kumar A, Kumar Katakam S, Cocola C, Pelucchi P, Graf M, Kiesel L, Reinbold R, Pavão MSG, Greve B, Götte M. The Heparan Sulfate Sulfotransferases HS2ST1 and HS3ST2 Are Novel Regulators of Breast Cancer Stem-Cell Properties. Front Cell Dev Biol 2020; 8:559554. [PMID: 33102470 PMCID: PMC7546021 DOI: 10.3389/fcell.2020.559554] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Heparan sulfate (HS) is a glycosaminoglycan found mainly in its protein-conjugated form at the cell surface and the extracellular matrix. Its high sulfation degree mediates functional interactions with positively charged amino acids in proteins. 2-O sulfation of iduronic acid and 3-O sulfation of glucosamine in HS are mediated by the sulfotransferases HS2ST and HS3ST, respectively, which are dysregulated in several cancers. Both sulfotransferases regulate breast cancer cell viability and invasion, but their role in cancer stem cells (CSCs) is unknown. Breast CSCs express characteristic markers such as CD44+/CD24−/low, CD133 and ALDH1 and are involved in tumor initiation, formation, and recurrence. We studied the influence of HS2ST1 and HS3ST2 overexpression on the CSC phenotype in breast cancer cell lines representative of the triple-negative (MDA-MB-231) and hormone-receptor positive subtype (MCF-7). The CD44+/CD24−/low phenotype was significantly reduced in MDA-MB-231 cells after overexpression of both enzymes, remaining unaltered in MCF-7 cells. ALDH1 activity was increased after HS2ST1 and HS3ST2 overexpression in MDA-MB-231 cells and reduced after HS2ST1 overexpression in MCF-7 cells. Colony and spheroid formation were increased after HS2ST1 and HS3ST2 overexpression in MCF-7 cells. Moreover, MDA-MB-231 cells overexpressing HS2ST1 formed more colonies and could not generate spheres. The phenotypic changes were associated with complex changes in the expression of the stemness-associated notch and Wnt-signaling pathways constituents, syndecans, heparanase and Sulf1. The results improve our understanding of breast CSC function and mark a subtype-specific impact of HS modifications on the CSC phenotype of triple-negative and hormone receptor positive breast cancer model cell lines.
Collapse
Affiliation(s)
- Felipe C O B Teixeira
- Instituto de Bioquímica Médica Leopoldo de Meis, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Archana Vijaya Kumar
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Sampath Kumar Katakam
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Cinzia Cocola
- Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche, Segrate, Italy
| | - Paride Pelucchi
- Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche, Segrate, Italy
| | - Monika Graf
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Rolland Reinbold
- Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche, Segrate, Italy
| | - Mauro S G Pavão
- Instituto de Bioquímica Médica Leopoldo de Meis, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Burkhard Greve
- Department of Radiotherapy and Radiooncology, University Hospital of Münster, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| |
Collapse
|