1
|
Le Pennec J, Makshakova O, Nevola P, Fouladkar F, Gout E, Machillot P, Friedel-Arboleas M, Picart C, Perez S, Vortkamp A, Vivès RR, Migliorini E. Glycosaminoglycans exhibit distinct interactions and signaling with BMP2 according to their nature and localization. Carbohydr Polym 2024; 341:122294. [PMID: 38876708 DOI: 10.1016/j.carbpol.2024.122294] [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: 03/25/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 06/16/2024]
Abstract
The role of glycosaminoglycans (GAGs) in modulating bone morphogenetic protein (BMP) signaling represents a recent and underexplored area. Conflicting reports suggest a dual effect: some indicate a positive influence, while others demonstrate a negative impact. This duality suggests that the localization of GAGs (either at the cell surface or within the extracellular matrix) or the specific type of GAG may dictate their signaling role. The precise sulfation patterns of heparan sulfate (HS) responsible for BMP2 binding remain elusive. BMP2 exhibits a preference for binding to HS over other GAGs. Using well-characterized biomaterials mimicking the extracellular matrix, our research reveals that HS promotes BMP2 signaling in the extracellular space, contrary to chondroitin sulfate (CS), which enhances BMP2 bioactivity at the cell surface. Further observations indicate that a central IdoA (2S)-GlcNS (6S) tri-sulfated motif within HS hexasaccharides enhances binding. Nevertheless, BMP2 exhibits a degree of adaptability to various HS sulfation types and sequences. Molecular dynamic simulations attribute this adaptability to the BMP2 N-terminal end flexibility. Our findings illustrate the complex interplay between GAGs and BMP signaling, highlighting the importance of localization and specific sulfation patterns. This understanding has implications for the development of biomaterials with tailored properties for therapeutic applications targeting BMP signaling pathways.
Collapse
Affiliation(s)
- Jean Le Pennec
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France
| | - Olga Makshakova
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, Synthetic Biology of Signalling Processes Lab, University of Freiburg, 79104 Freiburg, Germany
| | - Paola Nevola
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France; Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale, University of Naples Federico II, Napoli, Italy
| | - Farah Fouladkar
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France
| | - Evelyne Gout
- Univ. Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Paul Machillot
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France
| | | | - Catherine Picart
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France
| | - Serge Perez
- Univ. Grenoble Alpes, CNRS, Centre de Recherche sur les Macromolécules Végétales, Grenoble, France
| | - Andrea Vortkamp
- Developmental Biology, Centre for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | | | - Elisa Migliorini
- Univ. Grenoble Alpes, INSERM, CEA, CNRS, U1292 Biosanté, EMR 5000, Grenoble, France.
| |
Collapse
|
2
|
Mosby L, Bowen A, Hadjivasiliou Z. Morphogens in the evolution of size, shape and patterning. Development 2024; 151:dev202412. [PMID: 39302048 PMCID: PMC7616732 DOI: 10.1242/dev.202412] [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] [Indexed: 10/13/2024]
Abstract
Much of the striking diversity of life on Earth has arisen from variations in the way that the same molecules and networks operate during development to shape and pattern tissues and organs into different morphologies. However, we still understand very little about the potential for diversification exhibited by different, highly conserved mechanisms during evolution, or, conversely, the constraints that they place on evolution. With the aim of steering the field in new directions, we focus on morphogen-mediated patterning and growth as a case study to demonstrate how conserved developmental mechanisms can adapt during evolution to drive morphological diversification and optimise functionality, and to illustrate how evolution algorithms and computational tools can be used alongside experiments to provide insights into how these conserved mechanisms can evolve. We first introduce key conserved properties of morphogen-driven patterning mechanisms, before summarising comparative studies that exemplify how changes in the spatiotemporal expression and signalling levels of morphogens impact the diversification of organ size, shape and patterning in nature. Finally, we detail how theoretical frameworks can be used in conjunction with experiments to probe the role of morphogen-driven patterning mechanisms in evolution. We conclude that morphogen-mediated patterning is an excellent model system and offers a generally applicable framework to investigate the evolution of developmental mechanisms.
Collapse
Affiliation(s)
- L.S. Mosby
- The Francis Crick Institute: Mathematical and Physical Biology Laboratory, 1 Midland Road, London, NW1 1AT, UK
- University College London: Department of Physics and Astronomy, Gower Street, London, WC1E 6BT, UK
- London Centre for Nanotechnology, 19 Gordon Street, London, WC1H 0AH, UK
| | - A.E. Bowen
- The Francis Crick Institute: Mathematical and Physical Biology Laboratory, 1 Midland Road, London, NW1 1AT, UK
- University College London: Department of Physics and Astronomy, Gower Street, London, WC1E 6BT, UK
| | - Z. Hadjivasiliou
- The Francis Crick Institute: Mathematical and Physical Biology Laboratory, 1 Midland Road, London, NW1 1AT, UK
- University College London: Department of Physics and Astronomy, Gower Street, London, WC1E 6BT, UK
- London Centre for Nanotechnology, 19 Gordon Street, London, WC1H 0AH, UK
| |
Collapse
|
3
|
Yassaghi Y, Nazerian Y, Niazi F, Niknejad H. Advancements in cell-based therapies for thermal burn wounds: a comprehensive systematic review of clinical trials outcomes. Stem Cell Res Ther 2024; 15:277. [PMID: 39227861 PMCID: PMC11373270 DOI: 10.1186/s13287-024-03901-2] [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: 04/18/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Burn trauma is one of the major causes of morbidity and mortality worldwide. The standard management of burn wounds consists of early debridement, dressing changes, surgical management, and split-thickness skin autografts (STSGs). However, there are limitations for the standard management that inclines us to find alternative treatment approaches, such as innovative cell-based therapies. We aimed to systematically review the different aspects of cell-based treatment approaches for burn wounds in clinical trials. METHODS A systematic search through PubMed, Medline, Embase, and Cochrane Library databases was carried out using a combination of keywords, including "Cell transplantation", "Fibroblast", "Keratinocyte", "Melanocyte", or "Stem Cell" with "Burn", "Burn wound", or "Burn injury". Firstly, titles and abstracts of the studies existing in these databases until "February 2024" were screened. Then, the selected studies were read thoroughly, and considering the inclusion and exclusion criteria, final articles were included in this systematic review. Moreover, a manual search was performed through the reference lists of the included studies to minimize the risk of missing reports. RESULTS Overall, 30 clinical trials with 970 patients were included in our study. Considering the type of cells, six studies used keratinocytes, nine used fibroblasts, eight used combined keratinocytes and fibroblasts, one study used combined keratinocytes and melanocytes, five used combined keratinocytes and fibroblasts and melanocytes, and one study used mesenchymal stem cells (MSCs). Evaluation of the preparation type in these studies showed that cultured method was used in 25 trials, and non-cultured method in 5 trials. Also, the graft type of 17 trials was allogeneic, and of 13 other trials was autologous. CONCLUSIONS Our study showed that employing cell-based therapies for the treatment of burn wounds have significant results in clinical studies and are promising approaches that can be considered as alternative treatments in many cases. However, choosing appropriate cell-based treatment for each burn wound is essential and depends on the situation of each patient.
Collapse
Affiliation(s)
- Younes Yassaghi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yasaman Nazerian
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Feizollah Niazi
- Department of Plastic and Reconstructive Surgery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Selarka K, Shravage BV. Illuminating intercellular autophagy: A comprehensive review of cell non-autonomous autophagy. Biochem Biophys Res Commun 2024; 716:150024. [PMID: 38701555 DOI: 10.1016/j.bbrc.2024.150024] [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: 03/31/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024]
Abstract
Macro-autophagy (autophagy hereafter) is an evolutionarily conserved cellular process that has long been recognized as an intracellular mechanism for maintaining cellular homeostasis. It involves the formation of a membraned structure called the autophagosome, which carries cargo that includes toxic protein aggregates and dysfunctional organelles to the lysosome for degradation and recycling. Autophagy is primarily considered and studied as a cell-autonomous mechanism. However, recent studies have illuminated an underappreciated facet of autophagy, i.e., non-autonomously regulated autophagy. Non-autonomously regulated autophagy involves the degradation of autophagic components, including organelles, cargo, and signaling molecules, and is induced in neighboring cells by signals from primary adjacent or distant cells/tissues/organs. This review provides insight into the complex molecular mechanisms governing non-autonomously regulated autophagy, highlighting the dynamic interplay between cells within tissue/organ or distinct cell types in different tissues/organs. Emphasis is placed on modes of intercellular communication that include secreted molecules, including microRNAs, and their regulatory roles in orchestrating this phenomenon. Furthermore, we explore the multidimensional roles of non-autonomously regulated autophagy in various physiological contexts, spanning tissue development and aging, as well as its importance in diverse pathological conditions, including cancer and neurodegeneration. By studying the complexities of non-autonomously regulated autophagy, we hope to gain insights into the sophisticated intercellular dynamics within multicellular organisms, including mammals. These studies will uncover novel avenues for therapeutic intervention to modulate intercellular autophagic pathways in altered human physiology.
Collapse
Affiliation(s)
- Karan Selarka
- Developmental Biology Group, MACS-Agharkar Research Institute, Pune, India; Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Bhupendra V Shravage
- Developmental Biology Group, MACS-Agharkar Research Institute, Pune, India; Department of Biotechnology, Savitribai Phule Pune University, Pune, India; Department of Zoology, Savitribai Phule Pune University, Pune, India.
| |
Collapse
|
5
|
Zhao M, Taniguchi Y, Shimono C, Jonouchi T, Cheng Y, Shimizu Y, Nalbandian M, Yamamoto T, Nakagawa M, Sekiguchi K, Sakurai H. Heparan Sulfate Chain-Conjugated Laminin-E8 Fragments Advance Paraxial Mesodermal Differentiation Followed by High Myogenic Induction from hiPSCs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308306. [PMID: 38685581 PMCID: PMC11234437 DOI: 10.1002/advs.202308306] [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: 11/01/2023] [Revised: 03/26/2024] [Indexed: 05/02/2024]
Abstract
Human-induced pluripotent stem cells (hiPSCs) have great therapeutic potential. The cell source differentiated from hiPSCs requires xeno-free and robust methods for lineage-specific differentiation. Here, a system is described for differentiating hiPSCs on new generation laminin fragments (NGLFs), a recombinant form of a laminin E8 fragment conjugated to the heparan sulfate chains (HS) attachment domain of perlecan. Using NGLFs, hiPSCs are highly promoted to direct differentiation into a paraxial mesoderm state with high-efficiency muscle lineage generation. HS conjugation to the C-terminus of Laminin E8 fragments brings fibroblast growth factors (FGFs) bound to the HS close to the cell surface of hiPSCs, thereby facilitating stronger FGF signaling pathways stimulation and initiating HOX gene expression, which triggers the paraxial mesoderm differentiation of hiPSCs. This highly efficient differentiation system can provide a roadmap for paraxial mesoderm development and an infinite source of myocytes and muscle stem cells for disease modeling and regenerative medicine.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Clinical ApplicationCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
- Center for Medical EpigeneticsSchool of Basic Medical SciencesChongqing Medical University1 Yixueyuan Road, Yuzhong DistrictChongqing400016China
| | - Yukimasa Taniguchi
- Division of Matrixome Research and ApplicationInstitute for Protein ResearchOsaka University3‐2 Yamadaoka, SuitaOsaka565‐0871Japan
| | - Chisei Shimono
- Division of Matrixome Research and ApplicationInstitute for Protein ResearchOsaka University3‐2 Yamadaoka, SuitaOsaka565‐0871Japan
| | - Tatsuya Jonouchi
- Department of Clinical ApplicationCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| | - Yushen Cheng
- Department of Life Science FrontiersCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| | - Yasuhiro Shimizu
- Division of Matrixome Research and ApplicationInstitute for Protein ResearchOsaka University3‐2 Yamadaoka, SuitaOsaka565‐0871Japan
| | - Minas Nalbandian
- Department of Clinical ApplicationCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| | - Takuya Yamamoto
- Department of Life Science FrontiersCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| | - Masato Nakagawa
- Department of Life Science FrontiersCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| | - Kiyotoshi Sekiguchi
- Division of Matrixome Research and ApplicationInstitute for Protein ResearchOsaka University3‐2 Yamadaoka, SuitaOsaka565‐0871Japan
| | - Hidetoshi Sakurai
- Department of Clinical ApplicationCenter for iPS Cell Research and Application (CiRA)Kyoto University53 Shogoin‐Kawahara‐cho, Sakyo‐kuKyoto606‐8507Japan
| |
Collapse
|
6
|
Polewski L, Moon E, Zappe A, Götze M, Szekeres GP, Roth C, Pagel K. Ion Mobility Mass Spectrometry-Based Disaccharide Analysis of Glycosaminoglycans. Chemistry 2024; 30:e202400783. [PMID: 38629399 DOI: 10.1002/chem.202400783] [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: 02/26/2024] [Indexed: 05/24/2024]
Abstract
Glycosaminoglycans (GAGs) are linear and acidic polysaccharides. They are ubiquitous molecules, which are involved in a wide range of biological processes. Despite being structurally simple at first glance, with a repeating backbone of alternating hexuronic acid and hexosamine dimers, GAGs display a highly complex structure, which predominantly results from their heterogeneous sulfation patterns. The commonly applied method for compositional analysis of all GAGs is "disaccharide analysis." In this process, GAGs are enzymatically depolymerized into disaccharides, derivatized with a fluorescent label, and then analysed through liquid chromatography. The limiting factor in the high throughput analysis of GAG disaccharides is the time-consuming liquid chromatography. To address this limitation, we here utilized trapped ion mobility-mass spectrometry (TIM-MS) for the separation of isomeric GAG disaccharides, which reduces the measurement time from hours to a few minutes. A full set of disaccharides comprises twelve structures, with eight possessing isomers. Most disaccharides cannot be differentiated by TIM-MS in underivatized form. Therefore, we developed chemical modifications to reduce sample complexity and enhance differentiability. Quantification is performed using stable isotope labelled standards, which are easily available due to the nature of the performed modifications.
Collapse
Affiliation(s)
- Lukasz Polewski
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Altensteinstraße 23a, Germany
- Department of Molecular Physics, Fritz-Haber-Institut der Max-Planck-Gesellschaft, 14195, Berlin, Faradayweg 4-6, Germany
| | - Eunjin Moon
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Altensteinstraße 23a, Germany
| | - Andreas Zappe
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Altensteinstraße 23a, Germany
| | - Michael Götze
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Altensteinstraße 23a, Germany
- Department of Molecular Physics, Fritz-Haber-Institut der Max-Planck-Gesellschaft, 14195, Berlin, Faradayweg 4-6, Germany
| | - Gergo Peter Szekeres
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Altensteinstraße 23a, Germany
- Department of Molecular Physics, Fritz-Haber-Institut der Max-Planck-Gesellschaft, 14195, Berlin, Faradayweg 4-6, Germany
| | - Christian Roth
- Department of Biomolecular Systems, Max-Planck Institute of Colloids and Interfaces, 14195, Berlin, Arnimallee 22, Germany
| | - Kevin Pagel
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Altensteinstraße 23a, Germany
- Department of Molecular Physics, Fritz-Haber-Institut der Max-Planck-Gesellschaft, 14195, Berlin, Faradayweg 4-6, Germany
| |
Collapse
|
7
|
Gędaj A, Gregorczyk P, Żukowska D, Chorążewska A, Ciura K, Kalka M, Porębska N, Opaliński Ł. Glycosylation of FGF/FGFR: An underrated sweet code regulating cellular signaling programs. Cytokine Growth Factor Rev 2024; 77:39-55. [PMID: 38719671 DOI: 10.1016/j.cytogfr.2024.04.001] [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: 03/21/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/22/2024]
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) constitute plasma-membrane localized signaling hubs that transmit signals from the extracellular environment to the cell interior, governing pivotal cellular processes like motility, metabolism, differentiation, division and death. FGF/FGFR signaling is critical for human body development and homeostasis; dysregulation of FGF/FGFR units is observed in numerous developmental diseases and in about 10% of human cancers. Glycosylation is a highly abundant posttranslational modification that is critical for physiological and pathological functions of the cell. Glycosylation is also very common within FGF/FGFR signaling hubs. Vast majority of FGFs (15 out of 22 members) are N-glycosylated and few FGFs are O-glycosylated. Glycosylation is even more abundant within FGFRs; all FGFRs are heavily N-glycosylated in numerous positions within their extracellular domains. A growing number of studies points on the multiple roles of glycosylation in fine-tuning FGF/FGFR signaling. Glycosylation modifies secretion of FGFs, determines their stability and affects interaction with FGFRs and co-receptors. Glycosylation of FGFRs determines their intracellular sorting, constitutes autoinhibitory mechanism within FGFRs and adjusts FGF and co-receptor recognition. Sugar chains attached to FGFs and FGFRs constitute also a form of code that is differentially decrypted by extracellular lectins, galectins, which transform FGF/FGFR signaling at multiple levels. This review focuses on the identified functions of glycosylation within FGFs and FGFRs and discusses their relevance for the cell physiology in health and disease.
Collapse
Affiliation(s)
- Aleksandra Gędaj
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Paulina Gregorczyk
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Dominika Żukowska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Aleksandra Chorążewska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Krzysztof Ciura
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Marta Kalka
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Natalia Porębska
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Łukasz Opaliński
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland.
| |
Collapse
|
8
|
Liu Y, Chen H, Xiao L, Dong P, Ma Y, Zhou Y, Yang J, Bian B, Xie G, Chen L, Shen L. Notum enhances gastric cancer stem-like cell properties through upregulation of Sox2 by PI3K/AKT signaling pathway. Cell Oncol (Dordr) 2024; 47:463-480. [PMID: 37749430 PMCID: PMC11090966 DOI: 10.1007/s13402-023-00875-w] [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] [Accepted: 09/09/2023] [Indexed: 09/27/2023] Open
Abstract
PURPOSE Considerable evidence suggests that tumor cells with stemness features contribute to initiation, progression, recurrence of gastric cancer (GC) and resistance to therapy, but involvement of underlying regulators and mechanisms remain largely unclear. However, the clinical significance and biological function of Notum in GC tumor sphere formation and tumorigenesis remain unclear. METHODS Bioinformatics analysis, RT-qPCR, western blot and imunohistochemistry staining were applied to characterize Notum expression in GC specimens. The early diagnostic value of Notum was analyzed by logistic regression analysis method. Cancer stemness assays were used in Notum knockdown and overexpressing cells in vitro and in vivo. RNA-seq was employed to reveal the downstream effectors of Notum. RESULTS Notum is highly expressed in early stage of GC patients and stem-like GC cells. For discriminating the early-stage and advanced GC patients, the joint analysis had a better diagnostic value. Overexpression of Notum markedly increased stemness features of GC cells to promote tumor sphere formation and tumorigenesis. Conversely, Notum knockdown attenuated the stem-like cell properties in vitro and in vivo. Mechanically, Notum upregulates Sox2 through activating the PI3K/AKT signaling pathway. Notum inhibitor Caffeine exhibited a potent inhibitory effect on stemness features by impairing the PI3K/AKT signaling pathway activity and targeting Sox2. CONCLUSION Our findings confer a comprehensive and mechanistic function of Notum in GC tumor sphere formation and tumorigenesis that may provide a novel and promising target for early diagnosis and clinical therapy of GC.
Collapse
Affiliation(s)
- Yi Liu
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Hui Chen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Lanshu Xiao
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Ping Dong
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yanhui Ma
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yunlan Zhou
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Junyao Yang
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Bingxian Bian
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guohua Xie
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Lei Chen
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Lisong Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, 200240, China.
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
9
|
Le Pennec J, Picart C, Vivès RR, Migliorini E. Sweet but Challenging: Tackling the Complexity of GAGs with Engineered Tailor-Made Biomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312154. [PMID: 38011916 DOI: 10.1002/adma.202312154] [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: 11/14/2023] [Indexed: 11/29/2023]
Abstract
Glycosaminoglycans (GAGs) play a crucial role in tissue homeostasis by regulating the activity and diffusion of bioactive molecules. Incorporating GAGs into biomaterials has emerged as a widely adopted strategy in medical applications, owing to their biocompatibility and ability to control the release of bioactive molecules. Nevertheless, immobilized GAGs on biomaterials can elicit distinct cellular responses compared to their soluble forms, underscoring the need to understand the interactions between GAG and bioactive molecules within engineered functional biomaterials. By controlling critical parameters such as GAG type, density, and sulfation, it becomes possible to precisely delineate GAG functions within a biomaterial context and to better mimic specific tissue properties, enabling tailored design of GAG-based biomaterials for specific medical applications. However, this requires access to pure and well-characterized GAG compounds, which remains challenging. This review focuses on different strategies for producing well-defined GAGs and explores high-throughput approaches employed to investigate GAG-growth factor interactions and to quantify cellular responses on GAG-based biomaterials. These automated methods hold considerable promise for improving the understanding of the diverse functions of GAGs. In perspective, the scientific community is encouraged to adopt a rational approach in designing GAG-based biomaterials, taking into account the in vivo properties of the targeted tissue for medical applications.
Collapse
Affiliation(s)
- Jean Le Pennec
- U1292 Biosanté, INSERM, CEA, Univ. Grenoble Alpes, CNRS EMR 5000 Biomimetism and Regenerative Medicine, Grenoble, F-38054, France
| | - Catherine Picart
- U1292 Biosanté, INSERM, CEA, Univ. Grenoble Alpes, CNRS EMR 5000 Biomimetism and Regenerative Medicine, Grenoble, F-38054, France
| | | | - Elisa Migliorini
- U1292 Biosanté, INSERM, CEA, Univ. Grenoble Alpes, CNRS EMR 5000 Biomimetism and Regenerative Medicine, Grenoble, F-38054, France
| |
Collapse
|
10
|
Jiménez-Jiménez C, Grobe K, Guerrero I. Hedgehog on the Move: Glypican-Regulated Transport and Gradient Formation in Drosophila. Cells 2024; 13:418. [PMID: 38474382 PMCID: PMC10930589 DOI: 10.3390/cells13050418] [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: 01/30/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Glypicans (Glps) are a family of heparan sulphate proteoglycans that are attached to the outer plasma membrane leaflet of the producing cell by a glycosylphosphatidylinositol anchor. Glps are involved in the regulation of many signalling pathways, including those that regulate the activities of Wnts, Hedgehog (Hh), Fibroblast Growth Factors (FGFs), and Bone Morphogenetic Proteins (BMPs), among others. In the Hh-signalling pathway, Glps have been shown to be essential for ligand transport and the formation of Hh gradients over long distances, for the maintenance of Hh levels in the extracellular matrix, and for unimpaired ligand reception in distant recipient cells. Recently, two mechanistic models have been proposed to explain how Hh can form the signalling gradient and how Glps may contribute to it. In this review, we describe the structure, biochemistry, and metabolism of Glps and their interactions with different components of the Hh-signalling pathway that are important for the release, transport, and reception of Hh.
Collapse
Affiliation(s)
- Carlos Jiménez-Jiménez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Nicolás Cabrera 1, E-28049 Madrid, Spain;
| | - Kay Grobe
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstrasse 15, 48149 Münster, Germany
| | - Isabel Guerrero
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Nicolás Cabrera 1, E-28049 Madrid, Spain;
| |
Collapse
|
11
|
Alghazali R, Nugud A, El-Serafi A. Glycan Modifications as Regulators of Stem Cell Fate. BIOLOGY 2024; 13:76. [PMID: 38392295 PMCID: PMC10886185 DOI: 10.3390/biology13020076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024]
Abstract
Glycosylation is a process where proteins or lipids are modified with glycans. The presence of glycans determines the structure, stability, and localization of glycoproteins, thereby impacting various biological processes, including embryogenesis, intercellular communication, and disease progression. Glycans can influence stem cell behavior by modulating signaling molecules that govern the critical aspects of self-renewal and differentiation. Furthermore, being located at the cell surface, glycans are utilized as markers for stem cell pluripotency and differentiation state determination. This review aims to provide a comprehensive overview of the current literature, focusing on the effect of glycans on stem cells with a reflection on the application of synthetic glycans in directing stem cell differentiation. Additionally, this review will serve as a primer for researchers seeking a deeper understanding of how synthetic glycans can be used to control stem cell differentiation, which may help establish new approaches to guide stem cell differentiation into specific lineages. Ultimately, this knowledge can facilitate the identification of efficient strategies for advancing stem cell-based therapeutic interventions.
Collapse
Affiliation(s)
- Raghad Alghazali
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 58183 Linköping, Sweden
| | - Ahmed Nugud
- Clinical Sciences, University of Edinburgh, Edinburgh EH4 2XU, UK
- Gastroenterology, Hepatology & Nutrition, Sheikh Khalifa Medical City, Abu Dhabi 51900, United Arab Emirates
| | - Ahmed El-Serafi
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, 58183 Linköping, Sweden
- Department of Hand Surgery, Plastic Surgery and Burns, Linköping University, 58185 Linköping, Sweden
| |
Collapse
|
12
|
Afsar B, Afsar RE. The role of glycosaminoglycans in blood pressure regulation. Microcirculation 2023; 30:e12832. [PMID: 37794746 DOI: 10.1111/micc.12832] [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: 02/01/2023] [Revised: 05/06/2023] [Accepted: 09/23/2023] [Indexed: 10/06/2023]
Abstract
Essential hypertension (HT) is the global health problem and is a major risk factor for the development of cardiovascular and kidney disease. High salt intake has been associated with HT and impaired kidney sodium excretion is considered to be a major mechanism for the development of HT. Although kidney has a very important role in regulation of BP, this traditional view of BP regulation was challenged by recent findings suggesting that nonosmotic tissue sodium deposition is very important for BP regulation. This new paradigm indicates that sodium can be stored and deposited nonosmotically in the interstitium without water retention and without increased BP. One of the major determinants of this deposition is glycosaminoglycans (GAGs). By binding to GAGs found in the endothelial surface layer (ESL) which contains glycocalyx, sodium is osmotically inactivated and not induce concurrent water retention. Thus, GAGs has important function for homeostatic BP and sodium regulation. In the current review, we summarized the role of GAGs in ESL and BP regulation.
Collapse
Affiliation(s)
- Baris Afsar
- School of Medicine, Department of Nephrology, Suleyman Demirel University, Isparta, Turkey
| | - Rengin Elsurer Afsar
- School of Medicine, Department of Nephrology, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
13
|
Subedi S, Hwang HJ, Kang D, Mehta PK, Kim N, Park H, Lee JS, Lee KH. Development of peptide-based ratiometric fluorescent probe for sensing heparan sulfate and heparin in aqueous solutions at physiological pH and quantitative detection of heparan sulfate in live cells. Biosens Bioelectron 2023; 238:115595. [PMID: 37595478 DOI: 10.1016/j.bios.2023.115595] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/01/2023] [Accepted: 08/11/2023] [Indexed: 08/20/2023]
Abstract
Heparan sulfate (HS) plays a critical role in various biological processes as a vital component of the extracellular matrix. In this study, we synthesized three fluorescent probes (1-3) comprising Arg-rich peptides as HS receptors and a fluorophore capable of exhibiting red-shifted emissions upon aggregation. All three probes demonstrated ratiometric responses to HS and heparin in aqueous solutions. Remarkably, probe 3 exhibited a unique ratiometric response to HS in both aqueous solutions at physiological pH and HS proteoglycans on live cells. Probe 3 displayed exceptional sensing properties, including high biocompatibility, water solubility, visible light excitation, a large Stokes shift for ratiometric detection and remarkable selectivity and sensitivity for HS (with a low limit of detection: 720 pM). Binding mode studies unveiled the crucial role of charge interactions between probe 3 and negatively charged HS sugar units. Upon binding, the fluorophore segments of the probes overlapped, inducing green and red emission changes through restricted intramolecular rotation of the fluorophore moiety. Importantly, probe 3 was effectively employed to quantify the reduction of HS proteoglycan levels in live cells by inhibiting HS sulfation using siRNA and an inhibitor. It successfully detected decreased HS levels in cells treated with doxorubicin and irradiation, consistent with results obtained from western blot and immunofluorescence assays. This study presents the first ratiometric fluorescent probe capable of quantitatively detecting HS levels in aqueous solutions and live cells. The unique properties of peptide-based probe 3 make it a valuable tool for studying HS biology and potentially for diagnostic applications in various biological systems.
Collapse
Affiliation(s)
- Sumita Subedi
- Research Center for Controlling Intercellular Communication and Education and Research Center for Smart Energy Materials and Process, South Korea; Department of Chemistry and Chemical Engineering, South Korea
| | - Hyun Jung Hwang
- Research Center for Controlling Intercellular Communication and Education and Research Center for Smart Energy Materials and Process, South Korea
| | - Donghee Kang
- Research Center for Controlling Intercellular Communication and Education and Research Center for Smart Energy Materials and Process, South Korea; Program in Biomedical Science & Engineering, Inha University, Incheon, 402-751, South Korea
| | - Pramod Kumar Mehta
- Research Center for Controlling Intercellular Communication and Education and Research Center for Smart Energy Materials and Process, South Korea; Department of Chemistry and Chemical Engineering, South Korea
| | - Nayeon Kim
- Research Center for Controlling Intercellular Communication and Education and Research Center for Smart Energy Materials and Process, South Korea; Program in Biomedical Science & Engineering, Inha University, Incheon, 402-751, South Korea
| | - Hyojin Park
- Research Center for Controlling Intercellular Communication and Education and Research Center for Smart Energy Materials and Process, South Korea; Department of Chemistry and Chemical Engineering, South Korea
| | - Jae-Seon Lee
- Research Center for Controlling Intercellular Communication and Education and Research Center for Smart Energy Materials and Process, South Korea; Program in Biomedical Science & Engineering, Inha University, Incheon, 402-751, South Korea.
| | - Keun-Hyeung Lee
- Research Center for Controlling Intercellular Communication and Education and Research Center for Smart Energy Materials and Process, South Korea; Department of Chemistry and Chemical Engineering, South Korea.
| |
Collapse
|
14
|
Nicoletti A, Vitale F, Quero G, Paratore M, Fiorillo C, Negri M, Carlino A, Inzani F, Gasbarrini A, Alfieri S, Zileri Dal Verme L. Immunohistochemical Evaluation of the Expression of Specific Membrane Antigens in Patients with Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:4586. [PMID: 37760554 PMCID: PMC10526869 DOI: 10.3390/cancers15184586] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/26/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
(1) Background: Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies. The lack of validated disease biomarkers makes timely diagnosis challenging in most cases. Cell membrane and surface proteins play a crucial role in several routes of oncogenesis. The aim of this study was to evaluate the expression of six membrane antigens on PDAC (CA 19-9, mucin 1 and 4 (MUC1, MUC4), mesothelin (MSLN), Annexin A10 (ANXA10), Glypican-1 (GPC-1)) and their correlation with oncologic outcomes. (2) Methods: Immunohistochemical staining for CA 19.9, MUC1, MUC4, MSLN, ANXA10, and GPC-1 of surgical samples of 50 consecutive patients with PDAC was performed. Antigen expression for tumor, ductal, and acinar tissues was classified according to the histo-score (H-score) by two pathologists. (3) Results: Recurrence rate was 47% and 18 patients (36%) deceased (median follow-up 21.5 months). Immunostaining for CA 19-9 and MUC1 showed a significantly higher expression in the neoplastic tissue compared to non-tumor ductal and acinar tissues (p < 0.001). MUC4, MSLN, ANXA10, and GPC-1 were selectively expressed in the neoplastic tissue (p < 0.001). A CA 19-9 H-score value >270 was independently associated with a worse overall survival (p = 0.05) and disease-free survival (p = 0.05). (4) Conclusions: CA 19-9 and MUC1 are highly expressed in PDAC cells. The histological expression of CA 19-9 may predict prognosis. MUC4, MSLN, ANXA10, and GPC-1 are selectively expressed by neoplastic tissue and may represent a potential histological biomarker of disease.
Collapse
Affiliation(s)
- Alberto Nicoletti
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| | - Federica Vitale
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| | - Giuseppe Quero
- Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (G.Q.); (C.F.); (S.A.)
| | - Mattia Paratore
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| | - Claudio Fiorillo
- Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (G.Q.); (C.F.); (S.A.)
| | - Marcantonio Negri
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| | - Angela Carlino
- Anatomia Patologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.C.); (F.I.)
| | - Frediano Inzani
- Anatomia Patologica, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.C.); (F.I.)
| | - Antonio Gasbarrini
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| | - Sergio Alfieri
- Chirurgia Digestiva, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (G.Q.); (C.F.); (S.A.)
| | - Lorenzo Zileri Dal Verme
- Pancreas Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (A.N.); (F.V.); (M.P.); (M.N.); (L.Z.D.V.)
| |
Collapse
|
15
|
Farrugia BL, Melrose J. The Glycosaminoglycan Side Chains and Modular Core Proteins of Heparan Sulphate Proteoglycans and the Varied Ways They Provide Tissue Protection by Regulating Physiological Processes and Cellular Behaviour. Int J Mol Sci 2023; 24:14101. [PMID: 37762403 PMCID: PMC10531531 DOI: 10.3390/ijms241814101] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
This review examines the roles of HS-proteoglycans (HS-PGs) in general, and, in particular, perlecan and syndecan as representative examples and their interactive ligands, which regulate physiological processes and cellular behavior in health and disease. HS-PGs are essential for the functional properties of tissues both in development and in the extracellular matrix (ECM) remodeling that occurs in response to trauma or disease. HS-PGs interact with a biodiverse range of chemokines, chemokine receptors, protease inhibitors, and growth factors in immune regulation, inflammation, ECM stabilization, and tissue protection. Some cell regulatory proteoglycan receptors are dually modified hybrid HS/CS proteoglycans (betaglycan, CD47). Neurexins provide synaptic stabilization, plasticity, and specificity of interaction, promoting neurotransduction, neurogenesis, and differentiation. Ternary complexes of glypican-1 and Robbo-Slit neuroregulatory proteins direct axonogenesis and neural network formation. Specific neurexin-neuroligin complexes stabilize synaptic interactions and neural activity. Disruption in these interactions leads to neurological deficits in disorders of functional cognitive decline. Interactions with HS-PGs also promote or inhibit tumor development. Thus, HS-PGs have complex and diverse regulatory roles in the physiological processes that regulate cellular behavior and the functional properties of normal and pathological tissues. Specialized HS-PGs, such as the neurexins, pikachurin, and Eyes-shut, provide synaptic stabilization and specificity of neural transduction and also stabilize the axenome primary cilium of phototoreceptors and ribbon synapse interactions with bipolar neurons of retinal neural networks, which are essential in ocular vision. Pikachurin and Eyes-Shut interactions with an α-dystroglycan stabilize the photoreceptor synapse. Novel regulatory roles for HS-PGs controlling cell behavior and tissue function are expected to continue to be uncovered in this fascinating class of proteoglycan.
Collapse
Affiliation(s)
- Brooke L. Farrugia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Raymond Purves Laboratory of Bone and Joint Research, Kolling Institute of Medical Research, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Sydney Medical School (Northern), University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
16
|
Makarova N, Lekka M, Gnanachandran K, Sokolov I. Mechanical Way To Study Molecular Structure of Pericellular Layer. ACS APPLIED MATERIALS & INTERFACES 2023; 15:35962-35972. [PMID: 37489588 PMCID: PMC10401571 DOI: 10.1021/acsami.3c06341] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023]
Abstract
Atomic force microscopy (AFM) has been used to study the mechanical properties of cells, in particular, malignant cells. Softening of various cancer cells compared to their nonmalignant counterparts has been reported for various cell types. However, in most AFM studies, the pericellular layer was ignored. As was shown, it could substantially change the measured cell rigidity and miss important information on the physical properties of the pericellular layer. Here we take into account the pericellular layer by using the brush model to do the AFM indentation study of bladder epithelial bladder nonmalignant (HCV29) and cancerous (TCCSUP) cells. It allows us to measure not only the quasistatic Young's modulus of the cell body but also the physical properties of the pericellular layer (the equilibrium length and grafting density). We found that the inner pericellular brush was longer for cancer cells, but its grafting density was similar to that found for nonmalignant cells. The outer brush was much shorter and less dense for cancer cells. Furthermore, we demonstrate a method to convert the obtained physical properties of the pericellular layer into biochemical language better known to the cell biology community. It is done by using heparinase I and neuraminidase enzymatic treatments that remove specific molecular parts of the pericellular layer. The presented here approach can also be used to decipher the molecular composition of not only pericellular but also other molecular layers.
Collapse
Affiliation(s)
- Nadezda Makarova
- Department
of Mechanical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Małgorzata Lekka
- Department
of Biophysical Microstructures, Institute
of Nuclear Physics PAN, PL-31342 Kraków, Poland
| | - Kajangi Gnanachandran
- Department
of Biophysical Microstructures, Institute
of Nuclear Physics PAN, PL-31342 Kraków, Poland
| | - Igor Sokolov
- Department
of Mechanical Engineering, Tufts University, Medford, Massachusetts 02155, United States
- Department
of Physics, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
17
|
Hu S, Xia C, Zou H, Ren W, Liu L, Wang L, Kang Q, He K, Wang T, Zhang X. HS6ST1 overexpressed in cancer-associated fibroblast and inhibited cholangiocarcinoma progression. Dig Liver Dis 2023; 55:1114-1125. [PMID: 36586771 DOI: 10.1016/j.dld.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/11/2022] [Accepted: 12/07/2022] [Indexed: 01/02/2023]
Abstract
BACKGROUD Fibroblasts turn into cancer associated fibroblasts (CAFs) in the tumor microenvironment, which play an important role in tumor progression. However, the mechanism is unclear. AIMS To investigate the role of CAFs with HS6ST1-overexpression in cell migration and invasion effects. METHODS Human primary CAFs were isolated and identified from intrahepatic cholangiocarcinoma. mRNA profiles differences between CAFs and NFs were examined by using transcriptome sequencing. Using Transwell® migration assays, ICCA cells (RBE and HUCCT1) with NF-CM, CAF-CM, CAFsNC-CM, and CAFsHS6ST1-CM were analyzed. Immunohistochemical staining were used to analyze the expression of HS6ST1 in CAF in 152 patients with ICCA. Overall survival (OS) was compared based on CAF HS6ST1 expression were analysed. The relationship between clinicopathological parameters and survival was also examined. RESULTS Successfully isolated CAFs is positive staining with αSMA, FSP-1, FAP, and PDGFR-β. Transcriptome sequencing showed that differently expressed genes were enriched in the function of the extracellular matrix and chemokine signaling pathway. HS6ST1 is differentially expressed between CAFs and NFs, and associated with the migration and invasion of ICCA cells. Moreover, HS6ST1 positive expression of CAFs predicted unfavorable prognosis in patients with intrahepatic cholangiocarcinoma and showed correlation with the presence of lymph node metastasis. CONCLUSION HS6ST1 is new possibilities for targeting the CAFs to reduce cholangiocarcinoma growth and metastasis.
Collapse
Affiliation(s)
- Sheng Hu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Chuqi Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hao Zou
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Wenjun Ren
- Department of Thoracic Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lixin Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Lianmin Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Qiang Kang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Kai He
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China
| | - Tao Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China.
| | - Xiaowen Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, 374# Dianmian Avenue, Kunming, Yunnan 650101, China.
| |
Collapse
|
18
|
Guss EJ, Akbergenova Y, Cunningham KL, Littleton JT. Loss of the extracellular matrix protein Perlecan disrupts axonal and synaptic stability during Drosophila development. eLife 2023; 12:RP88273. [PMID: 37368474 PMCID: PMC10328508 DOI: 10.7554/elife.88273] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) form essential components of the extracellular matrix (ECM) and basement membrane (BM) and have both structural and signaling roles. Perlecan is a secreted ECM-localized HSPG that contributes to tissue integrity and cell-cell communication. Although a core component of the ECM, the role of Perlecan in neuronal structure and function is less understood. Here, we identify a role for Drosophila Perlecan in the maintenance of larval motoneuron axonal and synaptic stability. Loss of Perlecan causes alterations in the axonal cytoskeleton, followed by axonal breakage and synaptic retraction of neuromuscular junctions. These phenotypes are not prevented by blocking Wallerian degeneration and are independent of Perlecan's role in Wingless signaling. Expression of Perlecan solely in motoneurons cannot rescue synaptic retraction phenotypes. Similarly, removing Perlecan specifically from neurons, glia, or muscle does not cause synaptic retraction, indicating the protein is secreted from multiple cell types and functions non-cell autonomously. Within the peripheral nervous system, Perlecan predominantly localizes to the neural lamella, a specialized ECM surrounding nerve bundles. Indeed, the neural lamella is disrupted in the absence of Perlecan, with axons occasionally exiting their usual boundary in the nerve bundle. In addition, entire nerve bundles degenerate in a temporally coordinated manner across individual hemi-segments throughout larval development. These observations indicate disruption of neural lamella ECM function triggers axonal destabilization and synaptic retraction of motoneurons, revealing a role for Perlecan in axonal and synaptic integrity during nervous system development.
Collapse
Affiliation(s)
- Ellen J Guss
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Yulia Akbergenova
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - Karen L Cunningham
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| | - J Troy Littleton
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
19
|
Pham SH, Vuorinen SI, Arif KT, Griffiths LR, Okolicsanyi RK, Haupt LM. Syndecan-4 regulates the HER2-positive breast cancer cell proliferation cells via CK19/AKT signalling. Biochimie 2023; 207:49-61. [PMID: 36460206 DOI: 10.1016/j.biochi.2022.11.010] [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: 05/10/2022] [Revised: 10/27/2022] [Accepted: 11/18/2022] [Indexed: 12/02/2022]
Abstract
Despite the use of the highly specific anti-HER2 receptor (trastuzumab) therapy, HER2-positive breast cancers account for 20-30% of all breast cancer carcinomas, with HER2 status a challenge to treatment interventions. The heparan sulfate proteoglycans (HSPGs) are prominently expressed in the extracellular matrix (ECM), mediate breast cancer proliferation, development, and metastasis with most studies to date conducted in animal models. This study examined HSPGs in HER2-positive human breast cancer cell lines and their contribution to cancer cell proliferation. The study examined the cells following enhancement (via the addition of heparin) and knockdown (KD; using short interfering RNA, siRNA) of HSPG core proteins. The interaction of HSPG core proteins and AKT signalling molecules was examined to identify any influence of this signalling pathway on cancer cell proliferation. Our findings illustrated the HSPG syndecan-4 (SDC4) core protein significantly regulates cell proliferation with increased BC cell proliferation following heparin addition to cultures and decreased cell number following SDC4 KD. In addition, along with SDC4, significant changes in CK19/AKT signalling were identified as mediators of BC HER2-positive BC cell proliferation. This study provides evidence for a cell growth regulatory axis involving HSPGs/CK19 and AKT that represents a potential molecular target to prevent proliferation of HER2-positive breast cancer cells.
Collapse
Affiliation(s)
- Son H Pham
- Queensland University of Technology (QUT), Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, 60 Musk Ave., Kelvin Grove, Queensland, 4059, Australia
| | - Sofia I Vuorinen
- Queensland University of Technology (QUT), Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, 60 Musk Ave., Kelvin Grove, Queensland, 4059, Australia
| | - Km Taufiqul Arif
- Queensland University of Technology (QUT), Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, 60 Musk Ave., Kelvin Grove, Queensland, 4059, Australia
| | - Lyn R Griffiths
- Queensland University of Technology (QUT), Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, 60 Musk Ave., Kelvin Grove, Queensland, 4059, Australia
| | - Rachel K Okolicsanyi
- Queensland University of Technology (QUT), Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, 60 Musk Ave., Kelvin Grove, Queensland, 4059, Australia
| | - Larisa M Haupt
- Queensland University of Technology (QUT), Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, 60 Musk Ave., Kelvin Grove, Queensland, 4059, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Australia.
| |
Collapse
|
20
|
Iyer KS, Prabhakara C, Mayor S, Rao M. Cellular compartmentalisation and receptor promiscuity as a strategy for accurate and robust inference of position during morphogenesis. eLife 2023; 12:e79257. [PMID: 36877545 PMCID: PMC9988261 DOI: 10.7554/elife.79257] [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: 04/05/2022] [Accepted: 01/14/2023] [Indexed: 03/07/2023] Open
Abstract
Precise spatial patterning of cell fate during morphogenesis requires accurate inference of cellular position. In making such inferences from morphogen profiles, cells must contend with inherent stochasticity in morphogen production, transport, sensing and signalling. Motivated by the multitude of signalling mechanisms in various developmental contexts, we show how cells may utilise multiple tiers of processing (compartmentalisation) and parallel branches (multiple receptor types), together with feedback control, to bring about fidelity in morphogenetic decoding of their positions within a developing tissue. By simultaneously deploying specific and nonspecific receptors, cells achieve a more accurate and robust inference. We explore these ideas in the patterning of Drosophila melanogaster wing imaginal disc by Wingless morphogen signalling, where multiple endocytic pathways participate in decoding the morphogen gradient. The geometry of the inference landscape in the high dimensional space of parameters provides a measure for robustness and delineates stiff and sloppy directions. This distributed information processing at the scale of the cell highlights how local cell autonomous control facilitates global tissue scale design.
Collapse
Affiliation(s)
- Krishnan S Iyer
- Simons Center for the Study of Living Machines, National Center for Biological Sciences - TIFRBangaloreIndia
| | | | - Satyajit Mayor
- National Center for Biological Sciences - TIFRBangaloreIndia
| | - Madan Rao
- Simons Center for the Study of Living Machines, National Center for Biological Sciences - TIFRBangaloreIndia
| |
Collapse
|
21
|
Alvarez-Rodrigo I, Willnow D, Vincent JP. The logistics of Wnt production and delivery. Curr Top Dev Biol 2023; 153:1-60. [PMID: 36967191 DOI: 10.1016/bs.ctdb.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Wnts are secreted proteins that control stem cell maintenance, cell fate decisions, and growth during development and adult homeostasis. Wnts carry a post-translational modification not seen in any other secreted protein: during biosynthesis, they are appended with a palmitoleoyl moiety that is required for signaling but also impairs solubility and hence diffusion in the extracellular space. In some contexts, Wnts act only in a juxtacrine manner but there are also instances of long range action. Several proteins and processes ensure that active Wnts reach the appropriate target cells. Some, like Porcupine, Wntless, and Notum are dedicated to Wnt function; we describe their activities in molecular detail. We also outline how the cell infrastructure (secretory, endocytic, and retromer pathways) contribute to the progression of Wnts from production to delivery. We then address how Wnts spread in the extracellular space and form a signaling gradient despite carrying a hydrophobic moiety. We highlight particularly the role of lipid-binding Wnt interactors and heparan sulfate proteoglycans. Finally, we briefly discuss how evolution might have led to the emergence of this unusual signaling pathway.
Collapse
|
22
|
Chemistry and Function of Glycosaminoglycans in the Nervous System. ADVANCES IN NEUROBIOLOGY 2023; 29:117-162. [DOI: 10.1007/978-3-031-12390-0_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
23
|
Kushige H, Amano Y, Yagi H, Morisaku T, Kojima H, Satou A, Hamada KI, Kitagawa Y. Injectable extracellular matrix hydrogels contribute to native cell infiltration in a rat partial nephrectomy model. J Biomed Mater Res B Appl Biomater 2023; 111:184-193. [PMID: 36053744 DOI: 10.1002/jbm.b.35144] [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: 03/04/2022] [Revised: 06/07/2022] [Accepted: 07/25/2022] [Indexed: 11/11/2022]
Abstract
Decellularized extracellular matrix (dECM) hydrogels have cytocompatibility, and are currently being investigated for application in soft tissues as a material that promotes native cell infiltration and tissue reconstruction. A dECM hydrogel has broad potential for application in organs with complex structures or various tissue injury models. In this study, we investigated the practical application of a dECM hydrogel by injecting a kidney-derived dECM hydrogel into a rat partial nephrectomy model. The prepared dECM hydrogel was adjustable in viscosity to allow holding at the excision site, and after gelation, had an elastic modulus similar to that of kidney tissue. In addition, the migration of renal epithelial cells and vascular endothelial cells embedded in dECM hydrogels was observed in vitro. Four weeks after injection of the dECM hydrogel to the partial excision site of the kidneys, infiltration of renal tubular constituent cells and native cells with high proliferative activity, as well as angiogenesis, were observed inside the injected areas. This study is the first to show that dECM hydrogels can be applied to the kidney, one of the most complex structural organs and that they can function as a scaffold to induce angiogenesis and infiltration of organ-specific renal tubular constituent cells, providing fundamental insights for further application of dECM hydrogels.
Collapse
Affiliation(s)
- Hiroko Kushige
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan.,JSR-Keio University Medical and Chemical Innovation Center (JKiC), Keio University School of Medicine, Tokyo, Japan
| | - Yuki Amano
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corp, Tokyo, Japan
| | - Hiroshi Yagi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan.,JSR-Keio University Medical and Chemical Innovation Center (JKiC), Keio University School of Medicine, Tokyo, Japan
| | - Toshinori Morisaku
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan.,JSR-Keio University Medical and Chemical Innovation Center (JKiC), Keio University School of Medicine, Tokyo, Japan
| | - Hideaki Kojima
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan.,JSR-Keio University Medical and Chemical Innovation Center (JKiC), Keio University School of Medicine, Tokyo, Japan
| | - Akiko Satou
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corp, Tokyo, Japan
| | - Ken-Ichi Hamada
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corp, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan.,JSR-Keio University Medical and Chemical Innovation Center (JKiC), Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Otsuka T, Kan HM, Mason TD, Nair LS, Laurencin CT. Overexpression of NDST1 Attenuates Fibrotic Response in Murine Adipose-Derived Stem Cells. Stem Cells Dev 2022; 31:787-798. [PMID: 35920108 PMCID: PMC9836701 DOI: 10.1089/scd.2022.0053] [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: 02/16/2022] [Accepted: 08/01/2022] [Indexed: 01/22/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) hold tremendous potential for treating diseases and repairing damaged tissues. Heparan sulfate (HS) plays various roles in cellular signaling mechanisms. The importance of HS in stem cell function has been reported and well documented. However, there has been little progress in using HS for therapeutic purposes. We focused on one of the sulfotransferases, NDST1, which influences overall HS chain extent and sulfation pattern, with the expectation to enhance stem cell function by increasing the N-sulfation level. We herein performed transfections of a green fluorescent protein-vector control and NDST1-vector into mouse ADSCs to evaluate stem cell functions. Overexpression of NDST1 suppressed the osteogenic differentiation of ADSCs. There was no pronounced effect observed on the stemness, inflammatory gene expression, nor any noticeable effect in adipogenic and chondrogenic differentiation. Under the tumor necrosis factor-alpha stimulation, NDST1 overexpression induced several chemokine productions that attract neutrophils and macrophages. Finally, we identified an antifibrotic response in ADSCs overexpressing NDST1. This study provides a foundation for the evaluation of HS-related effects in ADSCs undergoing ex vivo gene manipulation.
Collapse
Affiliation(s)
- Takayoshi Otsuka
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health, Farmington, Connecticut, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, Connecticut, USA
| | - Ho-Man Kan
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health, Farmington, Connecticut, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, Connecticut, USA
| | - Timothy D. Mason
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health, Farmington, Connecticut, USA
| | - Lakshmi S. Nair
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health, Farmington, Connecticut, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, Connecticut, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Cato T. Laurencin
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health, Farmington, Connecticut, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, Connecticut, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, Connecticut, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, Connecticut, USA
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
25
|
Heparanase: A Novel Therapeutic Target for the Treatment of Atherosclerosis. Cells 2022; 11:cells11203198. [PMID: 36291066 PMCID: PMC9599978 DOI: 10.3390/cells11203198] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death and disability worldwide, and its management places a huge burden on healthcare systems through hospitalisation and treatment. Atherosclerosis is a chronic inflammatory disease of the arterial wall resulting in the formation of lipid-rich, fibrotic plaques under the subendothelium and is a key contributor to the development of CVD. As such, a detailed understanding of the mechanisms involved in the development of atherosclerosis is urgently required for more effective disease treatment and prevention strategies. Heparanase is the only mammalian enzyme known to cleave heparan sulfate of heparan sulfate proteoglycans, which is a key component of the extracellular matrix and basement membrane. By cleaving heparan sulfate, heparanase contributes to the regulation of numerous physiological and pathological processes such as wound healing, inflammation, tumour angiogenesis, and cell migration. Recent evidence suggests a multifactorial role for heparanase in atherosclerosis by promoting underlying inflammatory processes giving rise to plaque formation, as well as regulating lesion stability. This review provides an up-to-date overview of the role of heparanase in physiological and pathological processes with a focus on the emerging role of the enzyme in atherosclerosis.
Collapse
|
26
|
Takahashi I. Importance of Heparan Sulfate Proteoglycans in Pancreatic Islets and β-Cells. Int J Mol Sci 2022; 23:12082. [PMID: 36292936 PMCID: PMC9603760 DOI: 10.3390/ijms232012082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 09/20/2022] [Indexed: 12/15/2022] Open
Abstract
β-cells in the islets of Langerhans of the pancreas secrete insulin in response to the glucose concentration in the blood. When these pancreatic β-cells are damaged, diabetes develops through glucose intolerance caused by insufficient insulin secretion. High molecular weight polysaccharides, such as heparin and heparan sulfate (HS) proteoglycans, and HS-degrading enzymes, such as heparinase, participate in the protection, maintenance, and enhancement of the functions of pancreatic islets and β-cells, and the demand for studies on glycobiology within the field of diabetes research has increased. This review introduces the roles of complex glycoconjugates containing high molecular weight polysaccharides and their degrading enzymes in pancreatic islets and β-cells, including those obtained in studies conducted by us earlier. In addition, from the perspective of glycobiology, this study proposes the possibility of application to diabetes medicine.
Collapse
Affiliation(s)
- Iwao Takahashi
- Division of Molecular and Cellular Pharmacology, Department of Pathophysiology and Pharmacology, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Morioka 028-3694, Iwate, Japan
| |
Collapse
|
27
|
Predictive model for cytoneme guidance in Hedgehog signaling based on Ihog- Glypicans interaction. Nat Commun 2022; 13:5647. [PMID: 36163184 PMCID: PMC9512826 DOI: 10.1038/s41467-022-33262-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 09/09/2022] [Indexed: 11/28/2022] Open
Abstract
During embryonic development, cell-cell communication is crucial to coordinate cell behavior, especially in the generation of differentiation patterns via morphogen gradients. Morphogens are signaling molecules secreted by a source of cells that elicit concentration-dependent responses in target cells. For several morphogens, cell-cell contact via filopodia-like-structures (cytonemes) has been proposed as a mechanism for their gradient formation. Despite of the advances on cytoneme signaling, little is known about how cytonemes navigate through the extracellular matrix and how they orient to find their target. For the Hedgehog (Hh) signaling pathway in Drosophila, Hh co-receptor and adhesion protein Interference hedgehog (Ihog) and the glypicans Dally and Dally-like-protein (Dlp) interact affecting the cytoneme behavior. Here, we describe that differences in the cytoneme stabilization and orientation depend on the relative levels of Ihog and glypicans, suggesting a mechanism for cytoneme guidance. Furthermore, we have developed a mathematical model to study and corroborate this cytoneme guiding mechanism. Cytonemes are specialized filopodia-like structures known to be involved in signal transduction. Here they propose a new predictive model for cytoneme guidance in Hedgehog signaling, which is based on Ihog, Dally, and Dlp protein levels.
Collapse
|
28
|
GPC1 Is Associated with Poor Prognosis and Treg Infiltration in Colon Adenocarcinoma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:8209700. [PMID: 36158119 PMCID: PMC9492339 DOI: 10.1155/2022/8209700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/08/2022] [Indexed: 12/24/2022]
Abstract
Glypican-1 (GPC1) is a glycosylated protein recognized as a promising biomarker for cancer. Nonetheless, there have been few systematic studies on GPC1 in colon adenocarcinoma (COAD). We conducted bioinformatic analysis based on The Cancer Genome Atlas (TCGA) and used clinical samples to verify that GPC1 is overexpressed in colon adenocarcinoma. Kaplan-Meier analysis showed that higher GPC1 expression was associated with poor overall survival (OS). The Cox regression model further showed that GPC1 expression is an independent negative prognostic factor for COAD. Gene set enrichment analysis demonstrated that multiple oncogenic signaling pathways were differentially enriched in GPC1 high- versus low-expressing COAD tumors, including DNA methylation, G2/M damage checkpoint, and telomere dysfunction. We observed a positive correlation between GPC1 expression and immune cell infiltration, such as regulatory T cells (Tregs), macrophages, and mast cells, and immunohistochemistry of 50 COAD tissues revealed that GPC1 expression was positively associated with Treg enrichment. Our results provide a promising candidate gene to predict the prognosis of COAD and new insights into tumor immunity. Further research is required to validate these results.
Collapse
|
29
|
Sahan AZ, Baday M, Patel CB. Biomimetic Hydrogels in the Study of Cancer Mechanobiology: Overview, Biomedical Applications, and Future Perspectives. Gels 2022; 8:gels8080496. [PMID: 36005097 PMCID: PMC9407355 DOI: 10.3390/gels8080496] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/26/2022] [Accepted: 07/02/2022] [Indexed: 11/18/2022] Open
Abstract
Hydrogels are biocompatible polymers that are tunable to the system under study, allowing them to be widely used in medicine, bioprinting, tissue engineering, and biomechanics. Hydrogels are used to mimic the three-dimensional microenvironment of tissues, which is essential to understanding cell–cell interactions and intracellular signaling pathways (e.g., proliferation, apoptosis, growth, and survival). Emerging evidence suggests that the malignant properties of cancer cells depend on mechanical cues that arise from changes in their microenvironment. These mechanobiological cues include stiffness, shear stress, and pressure, and have an impact on cancer proliferation and invasion. The hydrogels can be tuned to simulate these mechanobiological tissue properties. Although interest in and research on the biomedical applications of hydrogels has increased in the past 25 years, there is still much to learn about the development of biomimetic hydrogels and their potential applications in biomedical and clinical settings. This review highlights the application of hydrogels in developing pre-clinical cancer models and their potential for translation to human disease with a focus on reviewing the utility of such models in studying glioblastoma progression.
Collapse
Affiliation(s)
- Ayse Z. Sahan
- Biomedical Sciences Graduate Program, Department of Pharmacology, School of Medicine, University California at San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Murat Baday
- Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Precision Health and Integrated Diagnostics Center, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Correspondence: (M.B.); (C.B.P.)
| | - Chirag B. Patel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
- Correspondence: (M.B.); (C.B.P.)
| |
Collapse
|
30
|
Sargison L, Smith RAA, Carnachan SM, Daines AM, Brackovic A, Kidgell JT, Nurcombe V, Cool SM, Sims IM, Hinkley SFR. Variability in the composition of porcine mucosal heparan sulfates. Carbohydr Polym 2022; 282:119081. [PMID: 35123736 DOI: 10.1016/j.carbpol.2021.119081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/16/2021] [Accepted: 12/28/2021] [Indexed: 11/18/2022]
Abstract
Commercial porcine intestinal mucosal heparan sulfate (HS) is a valuable material for research into its biological functions. As it is usually produced as a side-stream of pharmaceutical heparin manufacture, its chemical composition may vary from batch to batch. We analysed the composition and structure of nine batches of HS from the same manufacturer. Statistical analysis of the disaccharide compositions placed these batches in three categories: group A had high GlcNAc and GlcNS, and low GlcN typical of HS; group B had high GlcN and GlcNS, and low GlcNAc; group C had high di- and trisulfated, and low unsulfated and monosulfated disaccharide repeats. These batches could be placed in the same categories based on their 1H NMR spectra and molecular weights. Anticoagulant and growth factor binding activities of these HS batches did not fit within these same groups but were related to the proportions of more highly sulfated disaccharide repeats.
Collapse
Affiliation(s)
- Liam Sargison
- The Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand.
| | - Raymond A A Smith
- Institute of Molecular and Cell Biology (IMCB), Glycotherapeutics Group, Agency for Science, Technology and Research (A*STAR), A*STAR, 138673, Singapore.
| | - Susan M Carnachan
- The Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand.
| | - Alison M Daines
- The Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand.
| | - Amira Brackovic
- The Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand.
| | - Joel T Kidgell
- The Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand.
| | - Victor Nurcombe
- Institute of Molecular and Cell Biology (IMCB), Glycotherapeutics Group, Agency for Science, Technology and Research (A*STAR), A*STAR, 138673, Singapore
| | - Simon M Cool
- Institute of Molecular and Cell Biology (IMCB), Glycotherapeutics Group, Agency for Science, Technology and Research (A*STAR), A*STAR, 138673, Singapore.
| | - Ian M Sims
- The Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand.
| | - Simon F R Hinkley
- The Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand.
| |
Collapse
|
31
|
Song L, Oseid DE, Wells EA, Coaston T, Robinson AS. Heparan Sulfate Proteoglycans (HSPGs) Serve as the Mediator Between Monomeric Tau and Its Subsequent Intracellular ERK1/2 Pathway Activation. J Mol Neurosci 2022; 72:772-791. [PMID: 35040015 PMCID: PMC8763444 DOI: 10.1007/s12031-021-01943-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/06/2021] [Indexed: 12/15/2022]
Abstract
The conversion of soluble tau protein to insoluble, hyperphosphorylated neurofibrillary tangles (NFTs) is a major hallmark leading to neuronal death observed in neurodegenerative tauopathies. Unlike NFTs, the involvement of monomeric tau in the progression of tau pathology has been less investigated. Using live-cell confocal microscopy and flow cytometry, we demonstrate that soluble 0N4R monomers were rapidly endocytosed by SH-SY5Y and C6 glioma cells via actin-dependent macropinocytosis. Further, cellular endocytosis of monomeric tau has been demonstrated to be HSPG-dependent, as shown in C6 glial cells with genetic knockouts of xylosyltransferase-1-a key enzyme in HSPG synthesis-with a reduced level of tau uptake. Tau internalization subsequently triggers ERK1/2 activation and therefore, the upregulation of IL-6 and IL-1β. The role of ERK1/2 in regulating the levels of pro-inflammatory gene transcripts was confirmed by inhibiting the MEK-ERK1/2 signaling pathway, which led to the attenuated IL-6 and IL-1β expressions but not that of TNF-α. Moreover, as a key regulator of tau internalization, LRP1 (low-density lipoprotein receptor-related protein 1) levels were downregulated in response to monomeric tau added to C6 cells, while it was upregulated in HSPG-deficient cells, suggesting that the involvement of LRP1 in tau uptake depends on the presence of HSPGs on the cell surface. The subsequent LRP1 knockdown experiment we performed shows that LRP1 deficiency leads to an attenuated propensity for tau uptake and further elevated IL-6 gene expression. Collectively, our data suggest that tau has multiple extracellular binding partners that mediate its internalization through distinct mechanisms. Additionally, this study demonstrates the important role of both HSPGs and LRP1 in regulating cellular immune responses to tau protein monomers, providing a novel target for alleviating the neuroinflammatory environment before the formation of neurofibrillary tangles.
Collapse
Affiliation(s)
- Liqing Song
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Daniel E Oseid
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70118, USA
| | - Evan A Wells
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Troy Coaston
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70118, USA
| | - Anne S Robinson
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70118, USA.
| |
Collapse
|
32
|
Vang S, Cochran P, Sebastian Domingo J, Krick S, Barnes JW. The Glycobiology of Pulmonary Arterial Hypertension. Metabolites 2022; 12:metabo12040316. [PMID: 35448503 PMCID: PMC9026683 DOI: 10.3390/metabo12040316] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive pulmonary vascular disease of complex etiology. Cases of PAH that do not receive therapy after diagnosis have a low survival rate. Multiple reports have shown that idiopathic PAH, or IPAH, is associated with metabolic dysregulation including altered bioavailability of nitric oxide (NO) and dysregulated glucose metabolism. Multiple processes such as increased proliferation of pulmonary vascular cells, angiogenesis, apoptotic resistance, and vasoconstriction may be regulated by the metabolic changes demonstrated in PAH. Recent reports have underscored similarities between metabolic abnormalities in cancer and IPAH. In particular, increased glucose uptake and altered glucose utilization have been documented and have been linked to the aforementioned processes. We were the first to report a link between altered glucose metabolism and changes in glycosylation. Subsequent reports have highlighted similar findings, including a potential role for altered metabolism and aberrant glycosylation in IPAH pathogenesis. This review will detail research findings that demonstrate metabolic dysregulation in PAH with an emphasis on glycobiology. Furthermore, this report will illustrate the similarities in the pathobiology of PAH and cancer and highlight the novel findings that researchers have explored in the field.
Collapse
|
33
|
Yang R, Chen M, Zheng J, Li X, Zhang X. The Role of Heparin and Glycocalyx in Blood-Brain Barrier Dysfunction. Front Immunol 2022; 12:754141. [PMID: 34992593 PMCID: PMC8724024 DOI: 10.3389/fimmu.2021.754141] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
The blood-brain barrier (BBB) functions as a dynamic boundary that protects the central nervous system from blood and plays an important role in maintaining the homeostasis of the brain. Dysfunction of the BBB is a pathophysiological characteristic of multiple neurologic diseases. Glycocalyx covers the luminal side of vascular endothelial cells(ECs). Damage of glycocalyx leads to disruption of the BBB, while inhibiting glycocalyx degradation maintains BBB integrity. Heparin has been recognized as an anticoagulant and it protects endothelial glycocalyx from destruction. In this review, we summarize the role of glycocalyx in BBB formation and the therapeutic potency of heparin to provide a theoretical basis for the treatment of neurological diseases related to BBB breakdown.
Collapse
Affiliation(s)
- Rui Yang
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Mingming Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jiayin Zheng
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xin Li
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaojuan Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
34
|
Waghmare I, Page-McCaw A. Regulation of Wnt distribution and function by Drosophila glypicans. J Cell Sci 2022; 135:274233. [PMID: 35112708 PMCID: PMC8918805 DOI: 10.1242/jcs.259405] [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: 02/05/2023] Open
Abstract
The extracellular distribution of secreted Wnt proteins is crucial for their ability to induce a response in target cells at short and long ranges to ensure proper development. Wnt proteins are evolutionarily conserved ligands that are lipid-modified, and their hydrophobic nature interferes with their solubility in the hydrophilic extracellular environment. This raises the question of how Wnt proteins spread extracellularly despite their lipid modifications, which are essential for both their secretion and function. Seminal studies on Drosophila Wingless (Wg), a prototypical Wnt, have discovered multiple mechanisms by which Wnt proteins spread. A central theme emerges from these studies: the Wnt lipid moiety is shielded from the aqueous environment, allowing the ligands to spread and remain viable for signaling. Wnt distribution in vivo is primarily facilitated by glypicans, which are cell-surface heparan sulfate proteoglycans, and recent studies have further provided mechanistic insight into how glypicans facilitate Wnt distribution. In this Review, we discuss the many diverse mechanisms of Wnt distribution, with a particular focus on glypican-mediated mechanisms.
Collapse
|
35
|
Yamada K, Yoshida K. Multiple subcellular localizations and functions of protein kinase Cδ in liver cancer. World J Gastroenterol 2022; 28:188-198. [PMID: 35110944 PMCID: PMC8776529 DOI: 10.3748/wjg.v28.i2.188] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/25/2021] [Accepted: 12/31/2021] [Indexed: 02/06/2023] Open
Abstract
Protein kinase Cδ (PKCδ) is a member of the PKC family, and its implications have been reported in various biological and cancerous processes, including cell proliferation, cell death, tumor suppression, and tumor progression. In liver cancer cells, accumulating reports show the bi-functional regulation of PKCδ in cell death and survival. PKCδ function is defined by various factors, such as phosphorylation, catalytic domain cleavage, and subcellular localization. PKCδ has multiple intracellular distribution patterns, ranging from the cytosol to the nucleus. We recently found a unique extracellular localization of PKCδ in liver cancer and its growth factor-like function in liver cancer cells. In this review, we first discuss the structural features of PKCδ and then focus on the functional diversity of PKCδ based on its subcellular localization, such as the nucleus, cell surface, and extracellular space. These findings improve our knowledge of PKCδ involvement in the progression of liver cancer.
Collapse
Affiliation(s)
- Kohji Yamada
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Kiyotsugu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| |
Collapse
|
36
|
Berns ML, Habas R. Biochemical Assays to Detect Activation of Small GTPases Rho, Rac, and Cdc42 during Morphogenesis. Methods Mol Biol 2022; 2438:83-95. [PMID: 35147936 DOI: 10.1007/978-1-0716-2035-9_5] [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] [Indexed: 06/14/2023]
Abstract
Wnt/Frizzled (Fz) signaling controls developmental, physiological, and pathological processes through several distinct pathways. Wnt/Fz activation of the small GTPases Rho, Rac, and Cdc42, is one key mechanism that regulates cell polarity and migration during vertebrate gastrulation. In this chapter, we describe biochemical assays for detection of Wnt/Fz-mediated activation of Rho, Rac and Cdc42 in both mammalian cells and Xenopus embryo explants.
Collapse
Affiliation(s)
- Mark L Berns
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| | - Raymond Habas
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Griffiths SC, Schwab RA, El Omari K, Bishop B, Iverson EJ, Malinauskas T, Dubey R, Qian M, Covey DF, Gilbert RJC, Rohatgi R, Siebold C. Hedgehog-Interacting Protein is a multimodal antagonist of Hedgehog signalling. Nat Commun 2021; 12:7171. [PMID: 34887403 PMCID: PMC8660895 DOI: 10.1038/s41467-021-27475-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/19/2021] [Indexed: 01/20/2023] Open
Abstract
Hedgehog (HH) morphogen signalling, crucial for cell growth and tissue patterning in animals, is initiated by the binding of dually lipidated HH ligands to cell surface receptors. Hedgehog-Interacting Protein (HHIP), the only reported secreted inhibitor of Sonic Hedgehog (SHH) signalling, binds directly to SHH with high nanomolar affinity, sequestering SHH. Here, we report the structure of the HHIP N-terminal domain (HHIP-N) in complex with a glycosaminoglycan (GAG). HHIP-N displays a unique bipartite fold with a GAG-binding domain alongside a Cysteine Rich Domain (CRD). We show that HHIP-N is required to convey full HHIP inhibitory function, likely by interacting with the cholesterol moiety covalently linked to HH ligands, thereby preventing this SHH-attached cholesterol from binding to the HH receptor Patched (PTCH1). We also present the structure of the HHIP C-terminal domain in complex with the GAG heparin. Heparin can bind to both HHIP-N and HHIP-C, thereby inducing clustering at the cell surface and generating a high-avidity platform for SHH sequestration and inhibition. Our data suggest a multimodal mechanism, in which HHIP can bind two specific sites on the SHH morphogen, alongside multiple GAG interactions, to inhibit SHH signalling.
Collapse
Affiliation(s)
- Samuel C Griffiths
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Evotec (UK) Ltd., Milton Park, Abingdon, UK
| | - Rebekka A Schwab
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kamel El Omari
- Science Division, Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK
| | - Benjamin Bishop
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ellen J Iverson
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tomas Malinauskas
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Ramin Dubey
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mingxing Qian
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MI, USA
| | - Douglas F Covey
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MI, USA
| | - Robert J C Gilbert
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Rajat Rohatgi
- Departments of Biochemistry and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
38
|
Gao J, Huang X. Recent advances on glycosyltransferases involved in the biosynthesis of the proteoglycan linkage region. Adv Carbohydr Chem Biochem 2021; 80:95-119. [PMID: 34872657 DOI: 10.1016/bs.accb.2021.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Proteoglycans (PGs) are an essential family of glycoproteins, which can play roles in many important biological events including cell proliferation, cancer development, and pathogen infections. Proteoglycans consist of a core protein with one or multiple glycosaminoglycan (GAG) chains, which are covalently attached to serine residues of serine-glycine dipeptide within the core protein through a common tetrasaccharide linkage. In the past three decades, four key glycosyl transferases involved in the biosynthesis of PG linkage have been discovered and investigated. This review aims to provide an overview on progress made on these four enzymes, with foci on enzyme expression/purification, substrate specificity, activity determination, product characterization, and structure-activity relationship analysis.
Collapse
Affiliation(s)
- Jia Gao
- Department of Chemistry, Michigan State University, East Lansing, MI, United States; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, MI, United States; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States; Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
39
|
Hu B, Rodriguez JJ, Kakkerla Balaraju A, Gao Y, Nguyen NT, Steen H, Suhaib S, Chen S, Lin F. Glypican 4 mediates Wnt transport between germ layers via signaling filopodia. J Cell Biol 2021; 220:212673. [PMID: 34591076 PMCID: PMC8488972 DOI: 10.1083/jcb.202009082] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 07/18/2021] [Accepted: 09/08/2021] [Indexed: 12/31/2022] Open
Abstract
Glypicans influence signaling pathways by regulating morphogen trafficking and reception. However, the underlying mechanisms in vertebrates are poorly understood. In zebrafish, Glypican 4 (Gpc4) is required for convergence and extension (C&E) of both the mesoderm and endoderm. Here, we show that transgenic expression of GFP-Gpc4 in the endoderm of gpc4 mutants rescued C&E defects in all germ layers. The rescue of mesoderm was likely mediated by Wnt5b and Wnt11f2 and depended on signaling filopodia rather than on cleavage of the Gpc4 GPI anchor. Gpc4 bound both Wnt5b and Wnt11f2 and regulated formation of the filopodia that transport Wnt5b and Wnt11f2 to neighboring cells. Moreover, this rescue was suppressed by blocking signaling filopodia that extend from endodermal cells. Thus, GFP-Gpc4–labeled protrusions that emanated from endodermal cells transported Wnt5b and Wnt11f2 to other germ layers, rescuing the C&E defects caused by a gpc4 deficiency. Our study reveals a new mechanism that could explain in vivo morphogen distribution involving Gpc4.
Collapse
Affiliation(s)
- Bo Hu
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Juan J Rodriguez
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Anurag Kakkerla Balaraju
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Yuanyuan Gao
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Nhan T Nguyen
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Heston Steen
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Saeb Suhaib
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Songhai Chen
- Department of Neuroscience and Pharmacology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| | - Fang Lin
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, IA
| |
Collapse
|
40
|
Abstract
The extracellular matrix (ECM) is an architecture that supports the cells in our bodies and regulates various cell functions. The ECM is composed of many proteins and carbohydrates, and these molecules activate various intracellular signaling pathways orchestrated to decide cell fates. Therefore, it is not enough to study the role of single ECM molecules to understand the roles of the ECM in the regulation of cell functions; it is necessary to understand how the ECM, as an assembly of various molecules, regulates cell functions as a whole. For this purpose, in vitro ECM models mimicking native ECM are required. Here, a decellularization technique is presented to reconstitute native ECM in vitro. In this article, methods for preparing decellularized ECM (dECM) are described for use in tumor and stem cell biology. Additionally, a method for confirmation of decellularization and a dECM modification method are described. These dECM types will be useful for comprehensive studies of ECM roles. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Preparation of in vitro extracellular matrix (ECM) models mimicking native ECM in different malignant tumor tissues Basic Protocol 2: Preparation of in vitro ECM models mimicking native ECM surrounding myoblasts differentiating into myotubes at each myogenic stage Support Protocol 1: Confirmation of myogenic stages by myogenic stages by myogenic gene expression analysis Basic Protocol 3: Confirmation of cell removal Basic Protocol 4: Reduction of chondroitin sulfate chains in cultured cell-derived decellularized ECM Support Protocol 2: Quantification of chondroitin sulfate chain amounts in the decellularized ECM.
Collapse
Affiliation(s)
- Takashi Hoshiba
- Biotechnology Group, Tokyo Metropolitan Industrial Technology Research Institute, Aomi, Koto-ku, Tokyo, Japan
| |
Collapse
|
41
|
Wishart TFL, Lovicu FJ. An Atlas of Heparan Sulfate Proteoglycans in the Postnatal Rat Lens. Invest Ophthalmol Vis Sci 2021; 62:5. [PMID: 34730792 PMCID: PMC8572486 DOI: 10.1167/iovs.62.14.5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Purpose The arrangement of lens cells is regulated by ocular growth factors. Although the effects of these inductive molecules on lens cell behavior (proliferation, survival, and fiber differentiation) are well-characterized, the precise mechanisms underlying the regulation of growth factor-mediated signaling in lens remains elusive. Increasing evidence highlights the importance of heparan sulfate proteoglycans (HSPGs) for the signaling regulation of growth factors; however, the identity of the different lens HSPGs and the specific roles they play in lens biology are still unknown. Methods Semiquantitative real-time (RT)‐PCR and immunolabeling were used to characterize the spatial distribution of all known HSPG core proteins and their associated glycosaminoglycans (heparan and chondroitin sulfate) in the postnatal rat lens. Fibroblast growth factor (FGF)-2-treated lens epithelial explants, cultured in the presence of Surfen (an inhibitor of heparan sulfate [HS]-growth factor binding interactions) were used to investigate the requirement for HS in FGF-2-induced proliferation, fiber differentiation, and ERK1/2-signaling. Results The lens expresses all HSPGs. These HSPGs are differentially localized to distinct functional regions of the lens. In vitro, inhibition of HS-sulfation with Surfen blocked FGF-2-mediated ERK1/2-signaling associated with lens epithelial cell proliferation and fiber differentiation, highlighting that these cellular processes are dependent on HS. Conclusions These findings support a requirement for HSPGs in FGF-2 driven lens cell proliferation and fiber differentiation. The identification of specific HSPG core proteins in key functional lens regions, and the divergent expression patterns of closely related HSPGs, suggests that different HSPGs may differentially regulate growth factor signaling networks leading to specific biological events involved in lens growth and maintenance.
Collapse
Affiliation(s)
- Tayler F L Wishart
- School of Medical Sciences, The University of Sydney, New South Wales, Australia
| | - Frank J Lovicu
- School of Medical Sciences, The University of Sydney, New South Wales, Australia.,Save Sight Institute, The University of Sydney, New South Wales, Australia
| |
Collapse
|
42
|
Xyloside Derivatives as Molecular Tools to Selectively Inhibit Heparan Sulfate and Chondroitin Sulfate Proteoglycan Biosynthesis. Methods Mol Biol 2021. [PMID: 34626420 DOI: 10.1007/978-1-0716-1398-6_56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Glycosaminoglycan (GAG) side chains of proteoglycans are involved in a wide variety of developmental and pathophysiological functions. Similar to a gene knockout, the ability to inhibit GAG biosynthesis would allow us to examine the function of endogenous GAG chains. However, ubiquitously and irreversibly knocking out all GAG biosynthesis would cause multiple effects, making it difficult to attribute a specific biological role to a specific GAG structure in spatiotemporal manner. Reversible and selective inhibition of GAG biosynthesis would allow us to examine the importance of endogenous GAGs to specific cellular, tissue, or organ systems. In this chapter, we describe the chemical synthesis and biological evaluation of xyloside derivatives as selective inhibitors of heparan sulfate and chondroitin/dermatan sulfate proteoglycan biosynthesis.
Collapse
|
43
|
Takahashi I. Role of Heparan Sulfate Proteoglycans in Insulin-producing Pancreatic β-cell Function. TRENDS GLYCOSCI GLYC 2021. [DOI: 10.4052/tigg.2028.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Iwao Takahashi
- Department of Medical Biochemistry, School of Pharmacy, Iwate Medical University
| |
Collapse
|
44
|
Takahashi I. Role of Heparan Sulfate Proteoglycans in Insulin-producing Pancreatic β-cell Function. TRENDS GLYCOSCI GLYC 2021. [DOI: 10.4052/tigg.2028.1e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Iwao Takahashi
- Department of Medical Biochemistry, School of Pharmacy, Iwate Medical University
| |
Collapse
|
45
|
Luo X, Campbell NA, He L, O’Brien DR, Singer MS, Lemjabbar-Alaoui H, Ahn KS, Smoot R, Torbenson MS, Rosen SD, Roberts LR. Sulfatase 2 (SULF2) Monoclonal Antibody 5D5 Suppresses Human Cholangiocarcinoma Xenograft Growth Through Regulation of a SULF2-Platelet-Derived Growth Factor Receptor Beta-Yes-Associated Protein Signaling Axis. Hepatology 2021; 74:1411-1428. [PMID: 33735525 PMCID: PMC9075007 DOI: 10.1002/hep.31817] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIMS Existing therapeutic approaches to treat cholangiocarcinoma (CCA) have limited effectiveness, prompting further study to develop therapies for CCA. We report a mechanistic role for the heparan sulfate editing enzyme sulfatase 2 (SULF2) in CCA pathogenesis. APPROACH AND RESULTS In silico analysis revealed elevated SULF2 expression in human CCA samples, occurring partly through gain of SULF2 copy number. We examined the effects of knockdown or overexpression of SULF2 on tumor growth, chemoresistance, and signaling pathway activity in human CCA cell lines in vitro. Up-regulation of SULF2 in CCA leads to increased platelet-derived growth factor receptor beta (PDGFRβ)-Yes-associated protein (YAP) signaling activity, promoting tumor growth and chemotherapy resistance. To explore the utility of targeting SULF2 in the tumor microenvironment for CCA treatment, we tested an anti-SULF2 mouse monoclonal antibody, 5D5, in a mouse CCA xenograft model. Targeting SULF2 by monoclonal antibody 5D5 inhibited PDGFRβ-YAP signaling and tumor growth in the mouse xenograft model. CONCLUSIONS These results suggest that SULF2 monoclonal antibody 5D5 or related agents may be potentially promising therapeutic agents in CCA.
Collapse
Affiliation(s)
- Xin Luo
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States,Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nellie A. Campbell
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States
| | - Li He
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States,Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Daniel R. O’Brien
- Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States
| | - Mark S. Singer
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Hassan Lemjabbar-Alaoui
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Keun Soo Ahn
- Department of Surgery, Keimyung University School of Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Rory Smoot
- Department of Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States
| | - Michael S. Torbenson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States
| | - Steven D. Rosen
- Department of Anatomy, University of California San Francisco, San Francisco, California, USA
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States,Corresponding author: Lewis R Roberts, MB ChB, PhD, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States; Tel: +1-507-266-3239; Fax: +1-507-284-0762:
| |
Collapse
|
46
|
Aronitz EM, Kamermans BA, Duffy KR. Development of parvalbumin neurons and perineuronal nets in the visual cortex of normal and dark-exposed cats. J Comp Neurol 2021; 529:2827-2841. [PMID: 33576496 DOI: 10.1002/cne.25127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 11/06/2022]
Abstract
During development, the visual system maintains a high capacity for modification by expressing characteristics permissive for plasticity, enabling neural circuits to be refined by visual experience to achieve their mature form. This period is followed by the emergence of characteristics that stabilize the brain to consolidate for lifetime connections that were informed by experience. Attenuation of plasticity potential is thought to derive from an accumulation of plasticity-inhibiting characteristics that appear at ages beyond the peak of plasticity. Perineuronal nets (PNNs) are molecular aggregations that primarily surround fast-spiking inhibitory neurons called parvalbumin (PV) cells, which exhibit properties congruent with a plasticity inhibitor. In this study, we examined the development of PNNs and PV cells in the primary visual cortex of a highly visual mammal, and assessed the impact that 10 days of darkness had on both characteristics. Here, we show that labeling for PV expression emerges earlier and reaches adult levels sooner than PNNs. We also demonstrate that darkness, a condition known to enhance plasticity, significantly reduces the density of PNNs and the size of PV cell somata but does not alter the number of PV cells in the visual cortex. The darkness-induced reduction of PV cell size occurred irrespective of whether neurons were surrounded by a PNN, suggesting that PNNs have a restricted capacity to inhibit plasticity. Finally, we show that PV cells surrounded by a PNN were significantly larger than those without one, supporting the view that PNNs may mediate trophic support to the cells they surround.
Collapse
Affiliation(s)
- Elise M Aronitz
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Braden A Kamermans
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kevin R Duffy
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
47
|
Yang S, Zhang Y, Yang C, Wu X, El Oud SM, Chen R, Cai X, Wu XS, Lan G, Zheng X. Competitive coordination of the dual roles of the Hedgehog co-receptor in homophilic adhesion and signal reception. eLife 2021; 10:65770. [PMID: 34003115 PMCID: PMC8131103 DOI: 10.7554/elife.65770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/30/2021] [Indexed: 12/18/2022] Open
Abstract
Hedgehog (Hh) signaling patterns embryonic tissues and contributes to homeostasis in adults. In Drosophila, Hh transport and signaling are thought to occur along a specialized class of actin-rich filopodia, termed cytonemes. Here, we report that Interference hedgehog (Ihog) not only forms a Hh receptor complex with Patched to mediate intracellular signaling, but Ihog also engages in trans-homophilic binding leading to cytoneme stabilization in a manner independent of its role as the Hh receptor. Both functions of Ihog (trans-homophilic binding for cytoneme stabilization and Hh binding for ligand sensing) involve a heparin-binding site on the first fibronectin repeat of the extracellular domain. Thus, the Ihog-Ihog interaction and the Hh-Ihog interaction cannot occur simultaneously for a single Ihog molecule. By combining experimental data and mathematical modeling, we determined that Hh-Ihog heterophilic interaction dominates and Hh can disrupt and displace Ihog molecules involved in trans-homophilic binding. Consequently, we proposed that the weaker Ihog-Ihog trans interaction promotes and stabilizes direct membrane contacts along cytonemes and that, as the cytoneme encounters secreted Hh ligands, the ligands trigger release of Ihog from trans Ihog-Ihog complex enabling transport or internalization of the Hh ligand-Ihog-Patched -receptor complex. Thus, the seemingly incompatible functions of Ihog in homophilic adhesion and ligand binding cooperate to assist Hh transport and reception along the cytonemes.
Collapse
Affiliation(s)
- Shu Yang
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Ya Zhang
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Chuxuan Yang
- Department of Physics, George Washington University, Washington, United States
| | - Xuefeng Wu
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Sarah Maria El Oud
- Department of Physics, George Washington University, Washington, United States
| | - Rongfang Chen
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Xudong Cai
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| | - Xufeng S Wu
- Laboratory of Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, United States
| | - Ganhui Lan
- Department of Physics, George Washington University, Washington, United States
| | - Xiaoyan Zheng
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, United States.,GW Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, United States
| |
Collapse
|
48
|
Li H, Liu S, Wu S, Li L, Ge R, Cheng CY. Bioactive fragments of laminin and collagen chains: lesson from the testis. Reproduction 2021; 159:R111-R123. [PMID: 31581125 DOI: 10.1530/rep-19-0288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/03/2019] [Indexed: 12/11/2022]
Abstract
Recent studies have shown that the testis is producing several biologically active peptides, namely the F5- and the NC1-peptides from laminin-γ3 and collagen α3 (IV) chain, respectively, that promotes blood-testis barrier (BTB) remodeling and also elongated spermatid release at spermiation. Also the LG3/4/5 peptide from laminin-α2 chain promotes BTB integrity which is likely being used for the assembly of a 'new' BTB behind preleptotene spermatocytes under transport at the immunological barrier. These findings thus provide a new opportunity for investigators to better understand the biology of spermatogenesis. Herein, we briefly summarize the recent findings and provide a critical update. We also present a hypothetical model which could serve as the framework for studies in the years to come.
Collapse
Affiliation(s)
- Huitao Li
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Shiwen Liu
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Siwen Wu
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Linxi Li
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| | - Renshan Ge
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - C Yan Cheng
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, New York, New York, USA
| |
Collapse
|
49
|
Shin D, Nakamura M, Morishita Y, Eiraku M, Yamakawa T, Sasamura T, Akiyama M, Inaki M, Matsuno K. Collective nuclear behavior shapes bilateral nuclear symmetry for subsequent left-right asymmetric morphogenesis in Drosophila. Development 2021; 148:260539. [PMID: 34097729 PMCID: PMC8126412 DOI: 10.1242/dev.198507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/23/2021] [Indexed: 11/20/2022]
Abstract
Proper organ development often requires nuclei to move to a specific position within the cell. To determine how nuclear positioning affects left-right (LR) development in the Drosophila anterior midgut (AMG), we developed a surface-modeling method to measure and describe nuclear behavior at stages 13-14, captured in three-dimensional time-lapse movies. We describe the distinctive positioning and a novel collective nuclear behavior by which nuclei align LR symmetrically along the anterior-posterior axis in the visceral muscles that overlie the midgut and are responsible for the LR-asymmetric development of this organ. Wnt4 signaling is crucial for the collective behavior and proper positioning of the nuclei, as are myosin II and the LINC complex, without which the nuclei fail to align LR symmetrically. The LR-symmetric positioning of the nuclei is important for the subsequent LR-asymmetric development of the AMG. We propose that the bilaterally symmetrical positioning of these nuclei may be mechanically coupled with subsequent LR-asymmetric morphogenesis.
Collapse
Affiliation(s)
- Dongsun Shin
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Mitsutoshi Nakamura
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Yoshitaka Morishita
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Mototsugu Eiraku
- Department of Biosystems Science, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tomoko Yamakawa
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Takeshi Sasamura
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Masakazu Akiyama
- Meiji Institute for Advanced Study of Mathematical Sciences, Meiji University, 4-21-1 Nakano, Nakanoku, Tokyo 164-8525, Japan
| | - Mikiko Inaki
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Kenji Matsuno
- Department of Biological Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
50
|
Mii Y, Nakazato K, Pack CG, Ikeda T, Sako Y, Mochizuki A, Taira M, Takada S. Quantitative analyses reveal extracellular dynamics of Wnt ligands in Xenopus embryos. eLife 2021; 10:55108. [PMID: 33904408 PMCID: PMC8139832 DOI: 10.7554/elife.55108] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
The mechanism of intercellular transport of Wnt ligands is still a matter of debate. To better understand this issue, we examined the distribution and dynamics of Wnt8 in Xenopus embryos. While Venus-tagged Wnt8 was found on the surfaces of cells close to Wnt-producing cells, we also detected its dispersal over distances of 15 cell diameters. A combination of fluorescence correlation spectroscopy and quantitative imaging suggested that only a small proportion of Wnt8 ligands diffuses freely, whereas most Wnt8 molecules are bound to cell surfaces. Fluorescence decay after photoconversion showed that Wnt8 ligands bound on cell surfaces decrease exponentially, suggesting a dynamic exchange of bound forms of Wnt ligands. Mathematical modeling based on this exchange recapitulates a graded distribution of bound, but not free, Wnt ligands. Based on these results, we propose that Wnt distribution in tissues is controlled by a dynamic exchange of its abundant bound and rare free populations.
Collapse
Affiliation(s)
- Yusuke Mii
- National Institute for Basic Biology and Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan.,The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan.,Japan Science and Technology Agency (JST), PRESTO, Kawaguchi, Japan.,Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | | | - Chan-Gi Pack
- Cellular Informatics Laboratory, RIKEN, Wako, Japan.,ASAN Institute for Life Sciences, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Takafumi Ikeda
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Yasushi Sako
- Cellular Informatics Laboratory, RIKEN, Wako, Japan
| | - Atsushi Mochizuki
- Theoretical Biology Laboratory, RIKEN, Wako, Japan.,Laboratory of Mathematical Biology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Masanori Taira
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan.,Department of Biological Sciences, Faculty of Science and Engineering, Chuo University, Tokyo, Japan
| | - Shinji Takada
- National Institute for Basic Biology and Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan.,The Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| |
Collapse
|