1
|
Evans AD, Pournoori N, Saksala E, Oommen OP. Glycosaminoglycans' for brain health: Harnessing glycosaminoglycan based biomaterials for treating central nervous system diseases and in-vitro modeling. Biomaterials 2024; 309:122629. [PMID: 38797120 DOI: 10.1016/j.biomaterials.2024.122629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/06/2024] [Accepted: 05/19/2024] [Indexed: 05/29/2024]
Abstract
Dysfunction of the central nervous system (CNS) following traumatic brain injuries (TBI), spinal cord injuries (SCI), or strokes remains challenging to address using existing medications and cell-based therapies. Although therapeutic cell administration, such as stem cells and neuronal progenitor cells (NPCs), have shown promise in regenerative properties, they have failed to provide substantial benefits. However, the development of living cortical tissue engineered grafts, created by encapsulating these cells within an extracellular matrix (ECM) mimetic hydrogel scaffold, presents a promising functional replacement for damaged cortex in cases of stroke, SCI, and TBI. These grafts facilitate neural network repair and regeneration following CNS injuries. Given that natural glycosaminoglycans (GAGs) are a major constituent of the CNS, GAG-based hydrogels hold potential for the next generation of CNS healing therapies and in vitro modeling of CNS diseases. Brain-specific GAGs not only offer structural and biochemical signaling support to encapsulated neural cells but also modulate the inflammatory response in lesioned brain tissue, facilitating host integration and regeneration. This review briefly discusses different roles of GAGs and their related proteoglycan counterparts in healthy and diseases brain and explores current trends and advancements in GAG-based biomaterials for treating CNS injuries and modeling diseases. Additionally, it examines injectable, 3D bioprintable, and conductive GAG-based scaffolds, highlighting their clinical potential for in vitro modeling of patient-specific neural dysfunction and their ability to enhance CNS regeneration and repair following CNS injury in vivo.
Collapse
Affiliation(s)
- Austin D Evans
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland
| | - Negin Pournoori
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland
| | - Emmi Saksala
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland
| | - Oommen P Oommen
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland; School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK.
| |
Collapse
|
2
|
Swart M, Redpath AN, Ogbechi J, Cardenas R, Topping L, Compeer EB, Goddard M, Chanalaris A, Williams R, Brewer DS, Smart N, Monaco C, Troeberg L. The extracellular heparan sulfatase SULF2 limits myeloid IFNβ signaling and Th17 responses in inflammatory arthritis. Cell Mol Life Sci 2024; 81:350. [PMID: 39141086 PMCID: PMC11335274 DOI: 10.1007/s00018-024-05333-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 08/15/2024]
Abstract
Heparan sulfate (HS) proteoglycans are important regulators of cellular responses to soluble mediators such as chemokines, cytokines and growth factors. We profiled changes in expression of genes encoding HS core proteins, biosynthesis enzymes and modifiers during macrophage polarisation, and found that the most highly regulated gene was Sulf2, an extracellular HS 6-O-sulfatase that was markedly downregulated in response to pro-inflammatory stimuli. We then generated Sulf2+/- bone marrow chimeric mice and examined inflammatory responses in antigen-induced arthritis, as a model of rheumatoid arthritis. Resolution of inflammation was impaired in myeloid Sulf2+/- chimeras, with elevated joint swelling and increased abundance of pro-arthritic Th17 cells in synovial tissue. Transcriptomic and in vitro analyses indicated that Sulf2 deficiency increased type I interferon signaling in bone marrow-derived macrophages, leading to elevated expression of the Th17-inducing cytokine IL6. This establishes that dynamic remodeling of HS by Sulf2 limits type I interferon signaling in macrophages, and so protects against Th17-driven pathology.
Collapse
Affiliation(s)
- Maarten Swart
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Andia N Redpath
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, OX1 3PT, UK
| | - Joy Ogbechi
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Ryan Cardenas
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Rosalind Franklin Road, Norwich, NR4 7UQ, UK
| | - Louise Topping
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Ewoud B Compeer
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Michael Goddard
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Anastasios Chanalaris
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Richard Williams
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Daniel S Brewer
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Rosalind Franklin Road, Norwich, NR4 7UQ, UK
| | - Nicola Smart
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Oxford, OX1 3PT, UK
| | - Claudia Monaco
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK
| | - Linda Troeberg
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK.
- Centre for Metabolic Health, Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Rosalind Franklin Road, Norwich, NR4 7UQ, UK.
| |
Collapse
|
3
|
Saied-Santiago K, Baxter M, Mathiaparanam J, Granato M. Serotonin neuromodulation directs optic nerve regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607648. [PMID: 39185204 PMCID: PMC11343150 DOI: 10.1101/2024.08.12.607648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Optic nerve (ON) regeneration in mammalian systems is limited by an overshadowing dominance of inhibitory factors. This has severely hampered the identification of pro-regenerative pathways. Here, we take advantage of the regenerative capacity of larval zebrafish to identify pathways that promote ON regeneration. From a small molecule screen, we identified modulators of serotonin (5-HT) signaling that inhibit ON regeneration. We find several serotonin type-1 receptor genes are expressed in RGC neurons during regeneration and that inhibiting 5-HT1 receptors or components of the 5-HT pathway selectively impedes ON regeneration. We show that 5-HT1 receptor signaling is dispensable during ON development yet is critical for regenerating axons to emerge from the injury site. Blocking 5-HT receptors once ON axons have crossed the chiasm does not inhibit regeneration, suggesting a selective role for 5-HT receptor signaling early during ON regeneration. Finally, we show that agonist-mediated activation of 5-HT1 receptors leads to enhanced and ectopic axonal regrowth. Combined, our results provide evidence for mechanisms through which serotonin-dependent neuromodulation directs ON regeneration in vivo.
Collapse
Affiliation(s)
- Kristian Saied-Santiago
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - Melissa Baxter
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - Jaffna Mathiaparanam
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| |
Collapse
|
4
|
Solaimuthu B, Khatib A, Tanna M, Karmi A, Hayashi A, Abu Rmaileh A, Lichtenstein M, Takoe S, Jolly MK, Shaul YD. The exostosin glycosyltransferase 1/STAT3 axis is a driver of breast cancer aggressiveness. Proc Natl Acad Sci U S A 2024; 121:e2316733121. [PMID: 38215181 PMCID: PMC10801894 DOI: 10.1073/pnas.2316733121] [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: 10/10/2023] [Accepted: 12/07/2023] [Indexed: 01/14/2024] Open
Abstract
The epithelial-mesenchymal transition (EMT) program is crucial for transforming carcinoma cells into a partially mesenchymal state, enhancing their chemoresistance, migration, and metastasis. This shift in cell state is tightly regulated by cellular mechanisms that are not yet fully characterized. One intriguing EMT aspect is the rewiring of the proteoglycan landscape, particularly the induction of heparan sulfate proteoglycan (HSPG) biosynthesis. This proteoglycan functions as a co-receptor that accelerates cancer-associated signaling pathways through its negatively-charged residues. However, the precise mechanisms through which EMT governs HSPG biosynthesis and its role in cancer cell plasticity remain elusive. Here, we identified exostosin glycosyltransferase 1 (EXT1), a central enzyme in HSPG biosynthesis, to be selectively upregulated in aggressive tumor subtypes and cancer cell lines, and to function as a key player in breast cancer aggressiveness. Notably, ectopic expression of EXT1 in epithelial cells is sufficient to induce HSPG levels and the expression of known mesenchymal markers, subsequently enhancing EMT features, including cell migration, invasion, and tumor formation. Additionally, EXT1 loss in MDA-MB-231 cells inhibits their aggressiveness-associated traits such as migration, chemoresistance, tumor formation, and metastasis. Our findings reveal that EXT1, through its role in HSPG biosynthesis, governs signal transducer and activator of transcription 3 (STAT3) signaling, a known regulator of cancer cell aggressiveness. Collectively, we present the EXT1/HSPG/STAT3 axis as a central regulator of cancer cell plasticity that directly links proteoglycan synthesis to oncogenic signaling pathways.
Collapse
Affiliation(s)
- Balakrishnan Solaimuthu
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Anees Khatib
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Mayur Tanna
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Abdelrahman Karmi
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Arata Hayashi
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Areej Abu Rmaileh
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Michal Lichtenstein
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| | - Suranjana Takoe
- Department of Biological Sciences, Indian Institute of Science Education and Research, Berhampur760010, India
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bangalore560012, India
| | - Yoav David Shaul
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem9112001, Israel
| |
Collapse
|
5
|
Yang C, Xue Y, Duan Y, Mao C, Wan M. Extracellular vesicles and their engineering strategies, delivery systems, and biomedical applications. J Control Release 2024; 365:1089-1123. [PMID: 38065416 DOI: 10.1016/j.jconrel.2023.11.057] [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: 07/19/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 01/07/2024]
Abstract
Extracellular vesicles are nanoscale vesicles that can be secreted by all cell types, are intracellular in origin and have the same composition as their parent cells, play a key role in intercellular communication in organismal health and disease, and are now often used as biomarkers of disease and therapeutic agents in biomedical research. When injected locally or systemically, they have the ability to provide a variety of therapeutic effects, for example, regeneration of skin damage or restoration of cardiac function. However, direct injection of extracellular vesicles may result in their rapid clearance from the injection site.In order to maintain the biological activity of extracellular vesicles and to control the release of effective concentrations for better therapeutic efficacy during long-term disease treatment, the design of an optimized drug delivery system is necessary and different systems for the continuous delivery of extracellular vesicles have been developed. This paper first provides an overview of the biogenesis, composition and physiological function of extracellular vesicles, followed by a review of different strategies for extracellular vesicle isolation and methods for engineering extracellular vesicles. In addition, this paper reviews the latest extracellular vesicle delivery platforms such as micro-nanoparticles, injectable hydrogels, microneedles and scaffold patches. At the same time, the research progress and key cases of extracellular vesicle delivery systems in the field of biomedical therapeutics are described. Finally, the challenges and future trends of extracellular vesicle delivery are discussed.
Collapse
Affiliation(s)
- Chunhao Yang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yunxin Xue
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yu Duan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
6
|
Vlodavsky I, Kayal Y, Hilwi M, Soboh S, Sanderson RD, Ilan N. Heparanase-A single protein with multiple enzymatic and nonenzymatic functions. PROTEOGLYCAN RESEARCH 2023; 1:e6. [PMID: 37547889 PMCID: PMC10398610 DOI: 10.1002/pgr2.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 08/08/2023]
Abstract
Heparanase (Hpa1) is expressed by tumor cells and cells of the tumor microenvironment and functions extracellularly to remodel the extracellular matrix (ECM) and regulate the bioavailability of ECM-bound factors, augmenting, among other effects, gene transcription, autophagy, exosome formation, and heparan sulfate (HS) turnover. Much of the impact of heparanase on tumor progression is related to its function in mediating tumor-host crosstalk, priming the tumor microenvironment to better support tumor growth, metastasis, and chemoresistance. The enzyme appears to fulfill some normal functions associated, for example, with vesicular traffic, lysosomal-based secretion, autophagy, HS turnover, and gene transcription. It activates cells of the innate immune system, promotes the formation of exosomes and autophagosomes, and stimulates signal transduction pathways via enzymatic and nonenzymatic activities. These effects dynamically impact multiple regulatory pathways that together drive tumor growth, dissemination, and drug resistance as well as inflammatory responses. The emerging premise is that heparanase expressed by tumor cells, immune cells, endothelial cells, and other cells of the tumor microenvironment is a key regulator of the aggressive phenotype of cancer, an important contributor to the poor outcome of cancer patients and a valid target for therapy. So far, however, antiheparanase-based therapy has not been implemented in the clinic. Unlike heparanase, heparanase-2 (Hpa2), a close homolog of heparanase (Hpa1), does not undergo proteolytic processing and hence lacks intrinsic HS-degrading activity, the hallmark of heparanase. Hpa2 retains the capacity to bind heparin/HS and exhibits an even higher affinity towards HS than heparanase, thus competing for HS binding and inhibiting heparanase enzymatic activity. It appears that Hpa2 functions as a natural inhibitor of Hpa1 regulates the expression of selected genes that maintain tissue hemostasis and normal function, and plays a protective role against cancer and inflammation, together emphasizing the significance of maintaining a proper balance between Hpa1 and Hpa2.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Yasmin Kayal
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Maram Hilwi
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Soaad Soboh
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Ralph D. Sanderson
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Neta Ilan
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| |
Collapse
|
7
|
Whitefield C, Vo Y, Schwartz BD, Hepburn C, Ahmed FH, Onagi H, Banwell MG, Nelms K, Malins LR, Jackson CJ. Complex Inhibitory Mechanism of Glycomimetics with Heparanase. Biochemistry 2023. [PMID: 37368361 DOI: 10.1021/acs.biochem.3c00038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Heparanase (HPSE) is the only mammalian endo-β-glucuronidase known to catalyze the degradation of heparan sulfate. Dysfunction of HPSE activity has been linked to several disease states, resulting in HPSE becoming the target of numerous therapeutic programs, yet no drug has passed clinical trials to date. Pentosan polysulfate sodium (PPS) is a heterogeneous, FDA-approved drug for the treatment of interstitial cystitis and a known HPSE inhibitor. However, due to its heterogeneity, characterization of its mechanism of HPSE inhibition is challenging. Here, we show that inhibition of HPSE by PPS is complex, involving multiple overlapping binding events, each influenced by factors such as oligosaccharide length and inhibitor-induced changes in the protein secondary structure. The present work advances our molecular understanding of the inhibition of HPSE and will aid in the development of therapeutics for the treatment of a broad range of pathologies associated with enzyme dysfunction, including cancer, inflammatory disease, and viral infections.
Collapse
Affiliation(s)
- Cassidy Whitefield
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Yen Vo
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Brett D Schwartz
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Caryn Hepburn
- Waters Australia Pty Ltd, 38-46 South Street, Rydalmere, New South Wales 2116, Australia
| | - F Hafna Ahmed
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Hideki Onagi
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Martin G Banwell
- Institute for Advanced and Applied Chemical Synthesis, College of Pharmacy, Jinan University, Guangzhou, Guangdong 510632, China
| | - Keats Nelms
- Beta Therapeutics Pty. Ltd. Level 6, 121 Marcus Clarke Street, Canberra, Australian Capital Territory 2601, Australia
| | - Lara R Malins
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| | - Colin J Jackson
- Research School of Chemistry, Australian National University, Canberra, Australian Capital Territory 2601, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra, Australian Capital Territory 2601, Australia
| |
Collapse
|
8
|
Wishart TFL, Lovicu FJ. Heparan sulfate proteoglycans (HSPGs) of the ocular lens. Prog Retin Eye Res 2023; 93:101118. [PMID: 36068128 DOI: 10.1016/j.preteyeres.2022.101118] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022]
Abstract
Heparan sulfate proteoglycans (HSPGs) reside in most cells; on their surface, in the pericellular milieu and/or extracellular matrix. In the eye, HSPGs can orchestrate the activity of key signalling molecules found in the ocular environment that promote its development and homeostasis. To date, our understanding of the specific roles played by individual HSPG family members, and the heterogeneity of their associated sulfated HS chains, is in its infancy. The crystalline lens is a relatively simple and well characterised ocular tissue that provides an ideal stage to showcase and model the expression and unique roles of individual HSPGs. Individual HSPG core proteins are differentially localised to eye tissues in a temporal and spatial developmental- and cell-type specific manner, and their loss or functional disruption results in unique phenotypic outcomes for the lens, and other ocular tissues. More recent work has found that different HS sulfation enzymes are also presented in a cell- and tissue-specific manner, and that disruption of these different sulfation patterns affects specific HS-protein interactions. Not surprisingly, these sulfated HS chains have also been reported to be required for lens and eye development, with dysregulation of HS chain structure and function leading to pathogenesis and eye-related phenotypes. In the lens, HSPGs undergo significant and specific changes in expression and function that can drive pathology, or in some cases, promote tissue repair. As master signalling regulators, HSPGs may one day serve as valuable biomarkers, and even as putative targets for the development of novel therapeutics, not only for the eye but for many other systemic pathologies.
Collapse
Affiliation(s)
- Tayler F L Wishart
- Molecular and Cellular Biomedicine, School of Medical Sciences, The University of Sydney, NSW, Australia.
| | - Frank J Lovicu
- Molecular and Cellular Biomedicine, School of Medical Sciences, The University of Sydney, NSW, Australia; Save Sight Institute, The University of Sydney, NSW, Australia.
| |
Collapse
|
9
|
Spead O, Moreland T, Weaver CJ, Costa ID, Hegarty B, Kramer KL, Poulain FE. Teneurin trans-axonal signaling prunes topographically missorted axons. Cell Rep 2023; 42:112192. [PMID: 36857189 PMCID: PMC10131173 DOI: 10.1016/j.celrep.2023.112192] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 01/04/2023] [Accepted: 02/14/2023] [Indexed: 03/02/2023] Open
Abstract
Building precise neural circuits necessitates the elimination of axonal projections that have inaccurately formed during development. Although axonal pruning is a selective process, how it is initiated and controlled in vivo remains unclear. Here, we show that trans-axonal signaling mediated by the cell surface molecules Glypican-3, Teneurin-3, and Latrophilin-3 prunes misrouted retinal axons in the visual system. Retinotopic neuron transplantations revealed that pioneer ventral axons that elongate first along the optic tract instruct the pruning of dorsal axons that missort in that region. Glypican-3 and Teneurin-3 are both selectively expressed by ventral retinal ganglion cells and cooperate for correcting missorted dorsal axons. The adhesion G-protein-coupled receptor Latrophilin-3 signals along dorsal axons to initiate the elimination of topographic sorting errors. Altogether, our findings show an essential function for Glypican-3, Teneurin-3, and Latrophilin-3 in topographic tract organization and demonstrate that axonal pruning can be initiated by signaling among axons themselves.
Collapse
Affiliation(s)
- Olivia Spead
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Trevor Moreland
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Cory J Weaver
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Irene Dalla Costa
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Brianna Hegarty
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | | | - Fabienne E Poulain
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
10
|
Cortés E, Pak JS, Özkan E. Structure and evolution of neuronal wiring receptors and ligands. Dev Dyn 2023; 252:27-60. [PMID: 35727136 PMCID: PMC10084454 DOI: 10.1002/dvdy.512] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 01/04/2023] Open
Abstract
One of the fundamental properties of a neuronal circuit is the map of its connections. The cellular and developmental processes that allow for the growth of axons and dendrites, selection of synaptic targets, and formation of functional synapses use neuronal surface receptors and their interactions with other surface receptors, secreted ligands, and matrix molecules. Spatiotemporal regulation of the expression of these receptors and cues allows for specificity in the developmental pathways that wire stereotyped circuits. The families of molecules controlling axon guidance and synapse formation are generally conserved across animals, with some important exceptions, which have consequences for neuronal connectivity. Here, we summarize the distribution of such molecules across multiple taxa, with a focus on model organisms, evolutionary processes that led to the multitude of such molecules, and functional consequences for the diversification or loss of these receptors.
Collapse
Affiliation(s)
- Elena Cortés
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Joseph S Pak
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Engin Özkan
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois, USA.,The Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
11
|
de Boer C, Armstrong Z, Lit VAJ, Barash U, Ruijgrok G, Boyango I, Weitzenberg MM, Schröder SP, Sarris AJC, Meeuwenoord NJ, Bule P, Kayal Y, Ilan N, Codée JDC, Vlodavsky I, Overkleeft HS, Davies GJ, Wu L. Mechanism-based heparanase inhibitors reduce cancer metastasis in vivo. Proc Natl Acad Sci U S A 2022; 119:e2203167119. [PMID: 35881786 PMCID: PMC9351465 DOI: 10.1073/pnas.2203167119] [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: 02/25/2022] [Accepted: 06/22/2022] [Indexed: 01/30/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) mediate essential interactions throughout the extracellular matrix (ECM), providing signals that regulate cellular growth and development. Altered HSPG composition during tumorigenesis strongly aids cancer progression. Heparanase (HPSE) is the principal enzyme responsible for extracellular heparan sulfate catabolism and is markedly up-regulated in aggressive cancers. HPSE overactivity degrades HSPGs within the ECM, facilitating metastatic dissemination and releasing mitogens that drive cellular proliferation. Reducing extracellular HPSE activity reduces cancer growth, but few effective inhibitors are known, and none are clinically approved. Inspired by the natural glycosidase inhibitor cyclophellitol, we developed nanomolar mechanism-based, irreversible HPSE inhibitors that are effective within physiological environments. Application of cyclophellitol-derived HPSE inhibitors reduces cancer aggression in cellulo and significantly ameliorates murine metastasis. Mechanism-based irreversible HPSE inhibition is an unexplored anticancer strategy. We demonstrate the feasibility of such compounds to control pathological HPSE-driven malignancies.
Collapse
Affiliation(s)
- Casper de Boer
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Zachary Armstrong
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
- Current address: Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Vincent A. J. Lit
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Uri Barash
- Technion Integrated Cancer Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Gijs Ruijgrok
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Ilanit Boyango
- Technion Integrated Cancer Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Merle M. Weitzenberg
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Sybrin P. Schröder
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Alexi J. C. Sarris
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Nico J. Meeuwenoord
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Pedro Bule
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
- Current address: Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal
| | - Yasmine Kayal
- Technion Integrated Cancer Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Neta Ilan
- Technion Integrated Cancer Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Jeroen D. C. Codée
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Israel Vlodavsky
- Technion Integrated Cancer Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Herman S. Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| | - Gideon J. Davies
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| | - Liang Wu
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
- The Rosalind Franklin Institute, Harwell, OX11 0FA, UK
| |
Collapse
|
12
|
Rahman M, Ramirez‐Suarez NJ, Diaz‐Balzac CA, Bülow HE. Specific N-glycans regulate an extracellular adhesion complex during somatosensory dendrite patterning. EMBO Rep 2022; 23:e54163. [PMID: 35586945 PMCID: PMC9253746 DOI: 10.15252/embr.202154163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/13/2022] [Accepted: 04/22/2022] [Indexed: 09/19/2023] Open
Abstract
N-glycans are molecularly diverse sugars borne by over 70% of proteins transiting the secretory pathway and have been implicated in protein folding, stability, and localization. Mutations in genes important for N-glycosylation result in congenital disorders of glycosylation that are often associated with intellectual disability. Here, we show that structurally distinct N-glycans regulate an extracellular protein complex involved in the patterning of somatosensory dendrites in Caenorhabditis elegans. Specifically, aman-2/Golgi alpha-mannosidase II, a conserved key enzyme in the biosynthesis of specific N-glycans, regulates the activity of the Menorin adhesion complex without obviously affecting the protein stability and localization of its components. AMAN-2 functions cell-autonomously to allow for decoration of the neuronal transmembrane receptor DMA-1/LRR-TM with the correct set of high-mannose/hybrid/paucimannose N-glycans. Moreover, distinct types of N-glycans on specific N-glycosylation sites regulate DMA-1/LRR-TM receptor function, which, together with three other extracellular proteins, forms the Menorin adhesion complex. In summary, specific N-glycan structures regulate dendrite patterning by coordinating the activity of an extracellular adhesion complex, suggesting that the molecular diversity of N-glycans can contribute to developmental specificity in the nervous system.
Collapse
Affiliation(s)
- Maisha Rahman
- Department of GeneticsAlbert Einstein College of MedicineBronxNYUSA
- Dominick P. Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNYUSA
| | - Nelson J Ramirez‐Suarez
- Department of GeneticsAlbert Einstein College of MedicineBronxNYUSA
- Present address:
Institute of Science and Technology AustriaKlosterneuburgAustria
| | - Carlos A Diaz‐Balzac
- Department of GeneticsAlbert Einstein College of MedicineBronxNYUSA
- Present address:
University of RochesterRochesterNYUSA
| | - Hannes E Bülow
- Department of GeneticsAlbert Einstein College of MedicineBronxNYUSA
- Dominick P. Purpura Department of NeuroscienceAlbert Einstein College of MedicineBronxNYUSA
| |
Collapse
|
13
|
Regulation of biomineralization by proteoglycans: From mechanisms to application. Carbohydr Polym 2022; 294:119773. [DOI: 10.1016/j.carbpol.2022.119773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022]
|
14
|
Xie S, Zhang Q, Jiang L. Current Knowledge on Exosome Biogenesis, Cargo-Sorting Mechanism and Therapeutic Implications. MEMBRANES 2022; 12:498. [PMID: 35629824 PMCID: PMC9144303 DOI: 10.3390/membranes12050498] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 02/04/2023]
Abstract
Extracellular vesicles (EVs) are nanoscale membrane vesicles released by donor cells that can be taken up by recipient cells. The study of EVs has the potential to identify unknown cellular and molecular mechanisms in intercellular communication and disease. Exosomes, with an average diameter of ≈100 nanometers, are a subset of EVs. Different molecular families have been shown to be involved in the formation of exosomes and subsequent secretion of exosomes, which largely leads to the complexity of the form, structure and function of exosomes. In addition, because of their low immunogenicity and ability to transfer a variety of bioactive components to recipient cells, exosomes are regarded as effective drug delivery systems. This review summarizes the known mechanisms of exosomes biogenesis, cargo loading, exosomes release and bioengineering, which is of great importance for further exploration into the clinical applications of EVs.
Collapse
Affiliation(s)
- Shenmin Xie
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding & Reproduction, Ministry of Agriculture, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China; (S.X.); (Q.Z.)
| | - Qin Zhang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding & Reproduction, Ministry of Agriculture, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China; (S.X.); (Q.Z.)
- College of Animal Science and Technology, Shandong Agricultural University, Tai’an 271018, China
| | - Li Jiang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding & Reproduction, Ministry of Agriculture, College of Animal Science & Technology, China Agricultural University, Beijing 100193, China; (S.X.); (Q.Z.)
| |
Collapse
|
15
|
Shin WR, Park DY, Kim JH, Lee JP, Thai NQ, Oh IH, Sekhon SS, Choi W, Kim SY, Cho BK, Kim SC, Min J, Ahn JY, Kim YH. Structure based innovative approach to analyze aptaprobe-GPC3 complexes in hepatocellular carcinoma. J Nanobiotechnology 2022; 20:204. [PMID: 35477501 PMCID: PMC9044640 DOI: 10.1186/s12951-022-01391-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/21/2022] [Indexed: 02/07/2023] Open
Abstract
Background Glypican-3 (GPC3), a membrane-bound heparan sulfate proteoglycan, is a biomarker of hepatocellular carcinoma (HCC) progression. Aptamers specifically binding to target biomolecules have recently emerged as clinical disease diagnosis targets. Here, we describe 3D structure-based aptaprobe platforms for detecting GPC3, such as aptablotting, aptaprobe-based sandwich assay (ALISA), and aptaprobe-based imaging analysis. Results For preparing the aptaprobe–GPC3 platforms, we obtained 12 high affinity aptamer candidates (GPC3_1 to GPC3_12) that specifically bind to target GPC3 molecules. Structure-based molecular interactions identified distinct aptatopic residues responsible for binding to the paratopic nucleotide sequences (nt-paratope) of GPC3 aptaprobes. Sandwichable and overlapped aptaprobes were selected through structural analysis. The aptaprobe specificity for using in HCC diagnostics were verified through Aptablotting and ALISA. Moreover, aptaprobe-based imaging showed that the binding property of GPC3_3 and their GPC3 specificity were maintained in HCC xenograft models, which may indicate a new HCC imaging diagnosis. Conclusion Aptaprobe has the potential to be used as an affinity reagent to detect the target in vivo and in vitro diagnosing system. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01391-z.
Collapse
Affiliation(s)
- Woo-Ri Shin
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Dae-Young Park
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Ji Hun Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jin-Pyo Lee
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Nguyen Quang Thai
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - In-Hwan Oh
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Simranjeet Singh Sekhon
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Wooil Choi
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, Jeonju, 54896, Republic of Korea
| | - Sung Yeon Kim
- College of Pharmacy, Wonkwang University, Shinyoung-dong 344-2, Iksan, Jeonbuk, 570-749, Republic of Korea
| | - Byung-Kwan Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Sun Chang Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, Jeonju, 54896, Republic of Korea.
| | - Ji-Young Ahn
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea.
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
16
|
Houlton J, Zubkova OV, Clarkson AN. Recovery of Post-Stroke Spatial Memory and Thalamocortical Connectivity Following Novel Glycomimetic and rhBDNF Treatment. Int J Mol Sci 2022; 23:ijms23094817. [PMID: 35563207 PMCID: PMC9101131 DOI: 10.3390/ijms23094817] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 12/10/2022] Open
Abstract
Stroke-induced cognitive impairments remain of significant concern, with very few treatment options available. The involvement of glycosaminoglycans in neuroregenerative processes is becoming better understood and recent advancements in technology have allowed for cost-effective synthesis of novel glycomimetics. The current study evaluated the therapeutic potential of two novel glycomimetics, compound A and G, when administered systemically five-days post-photothrombotic stroke to the PFC. As glycosaminoglycans are thought to facilitate growth factor function, we also investigated the combination of our glycomimetics with intracerebral, recombinant human brain-derived neurotrophic factor (rhBDNF). C56BL/6J mice received sham or stroke surgery and experimental treatment (day-5), before undergoing the object location recognition task (OLRT). Four-weeks post-surgery, animals received prelimbic injections of the retrograde tracer cholera toxin B (CTB), before tissue was collected for quantification of thalamo-PFC connectivity and reactive astrogliosis. Compound A or G treatment alone modulated a degree of reactive astrogliosis yet did not influence spatial memory performance. Contrastingly, compound G+rhBDNF treatment significantly improved spatial memory, dampened reactive astrogliosis and limited stroke-induced loss of connectivity between the PFC and midline thalamus. As rhBDNF treatment had negligible effects, these findings support compound A acted synergistically to enhance rhBDNF to restrict secondary degeneration and facilitate functional recovery after PFC stroke.
Collapse
Affiliation(s)
- Josh Houlton
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin 9054, New Zealand;
| | - Olga V. Zubkova
- The Ferrier Research Institute, Gracefield Research Centre, Victoria University of Wellington, 69 Gracefield Road, Lower Hutt 5040, New Zealand;
| | - Andrew N. Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin 9054, New Zealand;
- Correspondence: ; Tel./Fax: +64-3-279-7326
| |
Collapse
|
17
|
Woodward AA, Taylor DM, Goldmuntz E, Mitchell LE, Agopian A, Moore JH, Urbanowicz RJ. Gene-Interaction-Sensitive enrichment analysis in congenital heart disease. BioData Min 2022; 15:4. [PMID: 35151364 PMCID: PMC8841104 DOI: 10.1186/s13040-022-00287-w] [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: 10/06/2021] [Accepted: 01/17/2022] [Indexed: 11/24/2022] Open
Abstract
Background Gene set enrichment analysis (GSEA) uses gene-level univariate associations to identify gene set-phenotype associations for hypothesis generation and interpretation. We propose that GSEA can be adapted to incorporate SNP and gene-level interactions. To this end, gene scores are derived by Relief-based feature importance algorithms that efficiently detect both univariate and interaction effects (MultiSURF) or exclusively interaction effects (MultiSURF*). We compare these interaction-sensitive GSEA approaches to traditional χ2 rankings in simulated genome-wide array data, and in a target and replication cohort of congenital heart disease patients with conotruncal defects (CTDs). Results In the simulation study and for both CTD datasets, both Relief-based approaches to GSEA captured more relevant and significant gene ontology terms compared to the univariate GSEA. Key terms and themes of interest include cell adhesion, migration, and signaling. A leading edge analysis highlighted semaphorins and their receptors, the Slit-Robo pathway, and other genes with roles in the secondary heart field and outflow tract development. Conclusions Our results indicate that interaction-sensitive approaches to enrichment analysis can improve upon traditional univariate GSEA. This approach replicated univariate findings and identified additional and more robust support for the role of the secondary heart field and cardiac neural crest cell migration in the development of CTDs. Supplementary Information The online version contains supplementary material available at (10.1186/s13040-022-00287-w).
Collapse
|
18
|
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
|
19
|
Garcia SA, Ng VY, Iwamoto M, Enomoto-Iwamoto M. Osteochondroma Pathogenesis: Mouse Models and Mechanistic Insights into Interactions with Retinoid Signaling. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2042-2051. [PMID: 34809786 PMCID: PMC8647428 DOI: 10.1016/j.ajpath.2021.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 07/27/2021] [Accepted: 08/12/2021] [Indexed: 05/02/2023]
Abstract
Osteochondromas are cartilage-capped tumors that arise near growing physes and are the most common benign bone tumor in children. Osteochondromas can lead to skeletal deformity, pain, loss of motion, and neurovascular compression. Currently, surgery is the only available treatment for symptomatic osteochondromas. Osteochondroma mouse models have been developed to understand the pathology and the origin of osteochondromas and develop therapeutic drugs. Several cartilage regulatory pathways have been implicated in the development of osteochondromas, such as bone morphogenetic protein, hedgehog, and WNT/β-catenin signaling. Retinoic acid receptor-γ is an important regulator of endochondral bone formation. Selective agonists for retinoic acid receptor-γ, such as palovarotene, have been investigated as drugs for inhibition of ectopic endochondral ossification, including osteochondromas. This review discusses the signaling pathways involved in osteochondroma pathogenesis and their possible interactions with the retinoid pathway.
Collapse
Affiliation(s)
- Sonia Arely Garcia
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Vincent Y Ng
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Masahiro Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
20
|
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
|
21
|
Revuelta J, Fraile I, Monterrey DT, Peña N, Benito-Arenas R, Bastida A, Fernández-Mayoralas A, García-Junceda E. Heparanized chitosans: towards the third generation of chitinous biomaterials. MATERIALS HORIZONS 2021; 8:2596-2614. [PMID: 34617543 DOI: 10.1039/d1mh00728a] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The functionalization of chitosans is an emerging research area in the design of solutions for a wide range of biomedical applications. In particular, the modification of chitosans to incorporate sulfate groups has generated great interest since they show structural similarity to heparin and heparan sulfates. Most of the biomedical applications of heparan sulfates are derived from their ability to bind different growth factors and other proteins, as through these interactions they can modulate the cellular response. This review aims to summarize the most recent advances in the synthesis, and structural and physicochemical characterization of heparanized chitosan, a remarkably interesting family of polysaccharides that have demonstrated the ability to mimic heparan sulfates as ligands for different proteins, thereby exerting their biological activity by mimicking the function of these glycosaminoglycans.
Collapse
Affiliation(s)
- Julia Revuelta
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Isabel Fraile
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Dianelis T Monterrey
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Nerea Peña
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Raúl Benito-Arenas
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Agatha Bastida
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Alfonso Fernández-Mayoralas
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| | - Eduardo García-Junceda
- BioGlycoChem Group, Departamento de Química Bio-Orgánica, Instituto de Química Orgánica General, CSIC (IQOG-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain.
| |
Collapse
|
22
|
Hayashi Y, Shibata A, Kamimura K, Kobayashi S. Heparan sulfate proteoglycan molecules, syndecan and perlecan, have distinct roles in the maintenance of Drosophila germline stem cells. Dev Growth Differ 2021; 63:295-305. [PMID: 34324711 DOI: 10.1111/dgd.12741] [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: 05/21/2021] [Revised: 07/04/2021] [Accepted: 07/27/2021] [Indexed: 11/29/2022]
Abstract
The Drosophila female germline stem cell (GSC) niche provides an excellent model for understanding the stem cell niche in vivo. The GSC niche is composed of stromal cells that provide growth factors for the maintenance of GSCs and the associated extracellular matrix (ECM). Although the function of stromal cells/growth factors has been well studied, the function of the ECM in the GSC niche is largely unknown. In this study, we investigated the function of syndecan and perlecan, molecules of the heparan sulfate proteoglycan (HSPG) family, as the main constituents of the ECM. We found that both of these genes were expressed in niche stromal cells, and knockdown of them in stromal cells decreased GSC number, indicating that these genes are important niche components. Interestingly, our genetic analysis revealed that the effects of syndecan and perlecan on the maintenance of GSC were distinct. While the knockdown of perlecan in the GSC niche increased the number of cystoblasts, a phenotype suggestive of delayed differentiation of GSCs, the same was not true in the context of syndecan. Notably, the overexpression of syndecan and perlecan did not cause an expansion of the GSC niche, opposing the results reported in the context of glypican, another HSPG gene. Altogether, our data suggest that HSPG genes contribute to the maintenance of GSCs through multiple mechanisms, such as the control of signal transduction, and ligand distribution/stabilization. Therefore, our study paves the way for a deeper understanding of the ECM functions in the stem cell niche.
Collapse
Affiliation(s)
- Yoshiki Hayashi
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| | - Arisa Shibata
- RIKEN Center for Sustainable Resource Science, Yokohama, Japan
| | - Keisuke Kamimura
- Tokyo Metropolitan Institute of Medical Science, Setagaya, Japan
| | - Satoru Kobayashi
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
23
|
Tortelote GG, Colón-Leyva M, Saifudeen Z. Metabolic programming of nephron progenitor cell fate. Pediatr Nephrol 2021; 36:2155-2164. [PMID: 33089379 PMCID: PMC10734399 DOI: 10.1007/s00467-020-04752-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/30/2020] [Accepted: 08/31/2020] [Indexed: 11/28/2022]
Abstract
Metabolic pathways are one of the first responses at the cellular level to maternal/fetal interface stressors. Studies have revealed the previously unrecognized contributions of intermediary metabolism to developmental programs. Here, we provide an overview of cellular metabolic pathways and the cues that modulate metabolic states. We discuss the developmental and physiological implications of metabolic reprogramming and the key role of metabolites in epigenetic and epiproteomic modifications during embryonic development and with respect to kidney development and nephrogenesis.
Collapse
Affiliation(s)
- Giovane G Tortelote
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue SL37, Room 5534, New Orleans, LA, 70112, USA
| | - Mariel Colón-Leyva
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue SL37, Room 5534, New Orleans, LA, 70112, USA
| | - Zubaida Saifudeen
- Department of Pediatrics, Tulane University School of Medicine, 1430 Tulane Avenue SL37, Room 5534, New Orleans, LA, 70112, USA.
| |
Collapse
|
24
|
Differences in MPS I and MPS II Disease Manifestations. Int J Mol Sci 2021; 22:ijms22157888. [PMID: 34360653 PMCID: PMC8345985 DOI: 10.3390/ijms22157888] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Mucopolysaccharidosis (MPS) type I and II are two closely related lysosomal storage diseases associated with disrupted glycosaminoglycan catabolism. In MPS II, the first step of degradation of heparan sulfate (HS) and dermatan sulfate (DS) is blocked by a deficiency in the lysosomal enzyme iduronate 2-sulfatase (IDS), while, in MPS I, blockage of the second step is caused by a deficiency in iduronidase (IDUA). The subsequent accumulation of HS and DS causes lysosomal hypertrophy and an increase in the number of lysosomes in cells, and impacts cellular functions, like cell adhesion, endocytosis, intracellular trafficking of different molecules, intracellular ionic balance, and inflammation. Characteristic phenotypical manifestations of both MPS I and II include skeletal disease, reflected in short stature, inguinal and umbilical hernias, hydrocephalus, hearing loss, coarse facial features, protruded abdomen with hepatosplenomegaly, and neurological involvement with varying functional concerns. However, a few manifestations are disease-specific, including corneal clouding in MPS I, epidermal manifestations in MPS II, and differences in the severity and nature of behavioral concerns. These phenotypic differences appear to be related to different ratios between DS and HS, and their sulfation levels. MPS I is characterized by higher DS/HS levels and lower sulfation levels, while HS levels dominate over DS levels in MPS II and sulfation levels are higher. The high presence of DS in the cornea and its involvement in the arrangement of collagen fibrils potentially causes corneal clouding to be prevalent in MPS I, but not in MPS II. The differences in neurological involvement may be due to the increased HS levels in MPS II, because of the involvement of HS in neuronal development. Current treatment options for patients with MPS II are often restricted to enzyme replacement therapy (ERT). While ERT has beneficial effects on respiratory and cardiopulmonary function and extends the lifespan of the patients, it does not significantly affect CNS manifestations, probably because the enzyme cannot pass the blood-brain barrier at sufficient levels. Many experimental therapies, therefore, aim at delivery of IDS to the CNS in an attempt to prevent neurocognitive decline in the patients.
Collapse
|
25
|
Moon S, Zhao YT. Spatial, temporal, and cell-type-specific expression profiles of genes encoding heparan sulfate biosynthesis enzymes and proteoglycan core proteins. Glycobiology 2021; 31:1308-1318. [PMID: 34132783 DOI: 10.1093/glycob/cwab054] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/28/2021] [Accepted: 06/09/2021] [Indexed: 12/22/2022] Open
Abstract
Heparan sulfate (HS) is a linear polysaccharide found in almost all animal cells and plays an important role in various biological processes. HS functions mainly via covalently binding to core proteins to form HS proteoglycans (HSPGs), which are heterogeneous in the lengths of the HS chain, the modifications on HS, and the core proteins. The molecular mechanisms underlying HSPG heterogeneity, although widely studied, are not yet fully defined. The expression profiles of HS biosynthesis enzymes and HSPG core proteins likely contribute to the HSPG heterogeneity, but these expression profiles remain poorly characterized. To investigate the expression profiles of genes encoding HS biosynthesis enzymes and HSPG core proteins, we systematically integrated the publicly available RNA sequencing data in mice. To reveal the spatial expression of these genes, we analyzed their expression in 21 mouse tissues. To reveal the temporal expression of these genes, we analyzed their expression at 17 time points during the mouse forebrain development. To determine the cell-type-specific expression of these genes, we obtained their expression profiles in 23 cell types in the mouse cerebral cortex by integrating single nucleus RNA sequencing data. Our findings demonstrate the spatial, temporal, and cell-type-specific expression of genes encoding HS biosynthesis enzymes and HSPG core proteins and represent a valuable resource to the HS research community.
Collapse
Affiliation(s)
- Sohyun Moon
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, 11568, USA
| | - Ying-Tao Zhao
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, 11568, USA
| |
Collapse
|
26
|
Cizeron M, Granger L, Bülow HE, Bessereau JL. Specific heparan sulfate modifications stabilize the synaptic organizer MADD-4/Punctin at C. elegans neuromuscular junctions. Genetics 2021; 218:6275221. [PMID: 33983408 DOI: 10.1093/genetics/iyab073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/16/2021] [Indexed: 01/01/2023] Open
Abstract
Heparan sulfate proteoglycans contribute to the structural organization of various neurochemical synapses. Depending on the system, their role involves either the core protein or the glycosaminoglycan chains. These linear sugar chains are extensively modified by heparan sulfate modification enzymes, resulting in highly diverse molecules. Specific modifications of glycosaminoglycan chains may thus contribute to a sugar code involved in synapse specificity. Caenorhabditis elegans is particularly useful to address this question because of the low level of genomic redundancy of these enzymes, as opposed to mammals. Here, we systematically mutated the genes encoding heparan sulfate modification enzymes in C. elegans and analyzed their impact on excitatory and inhibitory neuromuscular junctions. Using single chain antibodies that recognize different heparan sulfate modification patterns, we show in vivo that these two heparan sulfate epitopes are carried by the SDN-1 core protein, the unique C. elegans syndecan orthologue, at neuromuscular junctions. Intriguingly, these antibodies differentially bind to excitatory and inhibitory synapses, implying unique heparan sulfate modification patterns at different neuromuscular junctions. Moreover, while most enzymes are individually dispensable for proper organization of neuromuscular junctions, we show that 3-O-sulfation of SDN-1 is required to maintain wild-type levels of the extracellular matrix protein MADD-4/Punctin, a central synaptic organizer that defines the identity of excitatory and inhibitory synaptic domains at the plasma membrane of muscle cells.
Collapse
Affiliation(s)
- Mélissa Cizeron
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, 69008 Lyon, France
| | - Laure Granger
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, 69008 Lyon, France
| | - Hannes E Bülow
- Department of Genetics & Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Jean-Louis Bessereau
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, 69008 Lyon, France
| |
Collapse
|
27
|
Habermann FA, Kaltner H, Higuero AM, García Caballero G, Ludwig AK, C. Manning J, Abad-Rodríguez J, Gabius HJ. What Cyto- and Histochemistry Can Do to Crack the Sugar Code. Acta Histochem Cytochem 2021; 54:31-48. [PMID: 34012175 PMCID: PMC8116616 DOI: 10.1267/ahc.21-00017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
As letters form the vocabulary of a language, biochemical 'symbols' (the building blocks of oligo- and polymers) make writing molecular messages possible. Compared to nucleotides and amino acids, sugars have chemical properties that facilitate to reach an unsurpassed level of oligomer diversity. These glycans are a part of the ubiquitous cellular glycoconjugates. Cyto- and histochemically, the glycans' structural complexity is mapped by glycophenotyping of cells and tissues using receptors ('readers', thus called lectins), hereby revealing its dynamic spatiotemporal regulation: these data support the concept of a sugar code. When proceeding from work with plant (haem)agglutinins as such tools to the discovery of endogenous (tissue) lectins, it became clear that a broad panel of biological meanings can indeed be derived from the sugar-based vocabulary (the natural glycome incl. post-synthetic modifications) by glycan-lectin recognition in situ. As consequence, the immunocyto- and histochemical analysis of lectin expression is building a solid basis for the steps toward tracking down functional correlations, for example in processes leading to cell adhesion, apoptosis, autophagy or growth regulation as well as targeted delivery of glycoproteins. Introduction of labeled tissue lectins to glycan profiling assists this endeavor by detecting counterreceptor(s) in situ. Combining these tools and their applications strategically will help to take the trip toward the following long-range aim: to compile a dictionary for the glycan vocabulary that translates each message (oligosaccharide) into its bioresponse(s), that is to crack the sugar code.
Collapse
Affiliation(s)
- Felix A. Habermann
- Institute of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Veterinärstr. 13, 80539 Munich, Germany
| | - Herbert Kaltner
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Veterinärstr. 13, 80539 Munich, Germany
| | - Alonso M. Higuero
- Membrane and Axonal Repair Laboratory, National Hospital for Paraplegics (SESCAM), Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Gabriel García Caballero
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Veterinärstr. 13, 80539 Munich, Germany
| | - Anna-Kristin Ludwig
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Veterinärstr. 13, 80539 Munich, Germany
| | - Joachim C. Manning
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Veterinärstr. 13, 80539 Munich, Germany
| | - José Abad-Rodríguez
- Membrane and Axonal Repair Laboratory, National Hospital for Paraplegics (SESCAM), Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Hans-Joachim Gabius
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Veterinärstr. 13, 80539 Munich, Germany
| |
Collapse
|
28
|
A complement factor H homolog, heparan sulfation, and syndecan maintain inversin compartment boundaries in C. elegans cilia. Proc Natl Acad Sci U S A 2021; 118:2016698118. [PMID: 33859044 DOI: 10.1073/pnas.2016698118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness among the elderly. Canonical disease models suggest that defective interactions between complement factor H (CFH) and cell surface heparan sulfate (HS) result in increased alternative complement pathway activity, cytolytic damage, and tissue inflammation in the retina. Although these factors are thought to contribute to increased disease risk, multiple studies indicate that noncanonical mechanisms that result from defective CFH and HS interaction may contribute to the progression of AMD as well. A total of 60 ciliated sensory neurons in the nematode Caenorhabditis elegans detect chemical, olfactory, mechanical, and thermal cues in the environment. Here, we find that a C. elegans CFH homolog localizes on CEP mechanosensory neuron cilia where it has noncanonical roles in maintaining inversin/NPHP-2 within its namesake proximal compartment and preventing inversin/NPHP-2 accumulation in distal cilia compartments in aging adults. CFH localization and maintenance of inversin/NPHP-2 compartment integrity depend on the HS 3-O sulfotransferase HST-3.1 and the transmembrane proteoglycan syndecan/SDN-1. Defective inversin/NPHP-2 localization in mouse and human photoreceptors with CFH mutations indicates that these functions and interactions may be conserved in vertebrate sensory neurons, suggesting that previously unappreciated defects in cilia structure may contribute to the progressive photoreceptor dysfunction associated with CFH loss-of-function mutations in some AMD patients.
Collapse
|
29
|
Jud MC, Lowry J, Padilla T, Clifford E, Yang Y, Fennell F, Miller AK, Hamill D, Harvey AM, Avila-Zavala M, Shao H, Nguyen Tran N, Bao Z, Bowerman B. A genetic screen for temperature-sensitive morphogenesis-defective Caenorhabditis elegans mutants. G3-GENES GENOMES GENETICS 2021; 11:6169531. [PMID: 33713117 PMCID: PMC8133775 DOI: 10.1093/g3journal/jkab026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/18/2021] [Indexed: 11/21/2022]
Abstract
Morphogenesis involves coordinated cell migrations and cell shape changes that generate tissues and organs, and organize the body plan. Cell adhesion and the cytoskeleton are important for executing morphogenesis, but their regulation remains poorly understood. As genes required for embryonic morphogenesis may have earlier roles in development, temperature-sensitive embryonic-lethal mutations are useful tools for investigating this process. From a collection of ∼200 such Caenorhabditis elegans mutants, we have identified 17 that have highly penetrant embryonic morphogenesis defects after upshifts from the permissive to the restrictive temperature, just prior to the cell shape changes that mediate elongation of the ovoid embryo into a vermiform larva. Using whole genome sequencing, we identified the causal mutations in seven affected genes. These include three genes that have roles in producing the extracellular matrix, which is known to affect the morphogenesis of epithelial tissues in multicellular organisms: the rib-1 and rib-2 genes encode glycosyltransferases, and the emb-9 gene encodes a collagen subunit. We also used live imaging to characterize epidermal cell shape dynamics in one mutant, or1219ts, and observed cell elongation defects during dorsal intercalation and ventral enclosure that may be responsible for the body elongation defects. These results indicate that our screen has identified factors that influence morphogenesis and provides a platform for advancing our understanding of this fundamental biological process.
Collapse
Affiliation(s)
- Molly C Jud
- Institute of Molecular Biology, University of Oregon, Eugene, OR, 97402, USA
| | - Josh Lowry
- Institute of Molecular Biology, University of Oregon, Eugene, OR, 97402, USA
| | - Thalia Padilla
- Institute of Molecular Biology, University of Oregon, Eugene, OR, 97402, USA
| | - Erin Clifford
- Institute of Molecular Biology, University of Oregon, Eugene, OR, 97402, USA
| | - Yuqi Yang
- Institute of Molecular Biology, University of Oregon, Eugene, OR, 97402, USA
| | - Francesca Fennell
- Institute of Molecular Biology, University of Oregon, Eugene, OR, 97402, USA
| | - Alexander K Miller
- Institute of Molecular Biology, University of Oregon, Eugene, OR, 97402, USA
| | - Danielle Hamill
- Department of Zoology, Ohio Wesleyan University, Delaware, OH, 43015, USA
| | - Austin M Harvey
- Institute of Molecular Biology, University of Oregon, Eugene, OR, 97402, USA
| | - Martha Avila-Zavala
- Institute of Molecular Biology, University of Oregon, Eugene, OR, 97402, USA
| | - Hong Shao
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Nhan Nguyen Tran
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Zhirong Bao
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, USA
| | - Bruce Bowerman
- Institute of Molecular Biology, University of Oregon, Eugene, OR, 97402, USA
| |
Collapse
|
30
|
Luong H, Singh S, Patil M, Krishnamurthy P. Cardiac glycosaminoglycans and structural alterations during chronic stress-induced depression-like behavior in mice. Am J Physiol Heart Circ Physiol 2021; 320:H2044-H2057. [PMID: 33834865 DOI: 10.1152/ajpheart.00635.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Major depressive disorder (MDD) is an independent risk factor for cardiovascular disease (CVD) and its complications; however, causal mechanisms remain unclear. In the present study, we investigate cardiac structural and functional alterations and associated changes in myocardial glycosaminoglycans (GAGs) disaccharide profile in mice that exhibit depression-like behavior. Mice were assigned to the chronic mild stress (CMS) group and nonstress control group (CT). The CMS group was exposed to a series of mild, unpredictable stressors for 7 wk. Mice in the CMS group show a significant decrease in protein expression of hippocampal brain-derived neurotrophic factor (BDNF) and exhibit depression-like behavioral changes, such as learned helplessness and decreased exploration behavior, as compared with the control group. Although cardiac function remained unchanged between the groups, echocardiography analysis showed slightly increased left ventricular wall thickness in the CMS group. Furthermore, the CMS group shows an increase in cardiomyocyte cross-sectional area and an associated decrease in BDNF protein expression and increase in IL-6 mRNA expression, when compared with control mice. GAG disaccharide analysis of the left ventricles of the CMS and CT mice revealed an elevation in heparan (HS) and chondroitin sulfate (CS) content in the CMS hearts (35.3% and 17.9%, respectively, vs. control group). Furthermore, we also observed that unsulfated or monosulfated disaccharides were the most abundant units; however, we did not find any significant difference in mole percent or sulfation pattern of HS/CS disaccharides between the groups. The current investigation highlights a need for further research to explore the relationship between cardiac GAGs biology and myocardial remodeling as a causal mechanism that underlie cardiovascular complications in patients with MDD.NEW & NOTEWORTHY Comorbidity between depression and CVD is well established, whereas its etiology, especially the role of nonfibrous components (proteoglycans/GAGs) of the extracellular matrix, is unexplored. To the best of our knowledge, this is the first study to characterize cardiac proteoglycan/glycosaminoglycan profile in response to depression-like behavioral changes in mice. We observed that chronic mild stress (CMS)-induced depression-like behavior and alterations in glycosaminoglycan profile were associated with structural changes in the heart.
Collapse
Affiliation(s)
- Hien Luong
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sarojini Singh
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mallikarjun Patil
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Prasanna Krishnamurthy
- Department of Biomedical Engineering, Schools of Medicine and Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
31
|
Peall IW, Okolicsanyi RK, Griffiths LR, Haupt LM. Three-Dimensional Human Neural Stem Cell Models to Mimic Heparan Sulfate Proteoglycans and the Neural Niche. Semin Thromb Hemost 2021; 47:308-315. [PMID: 33794554 DOI: 10.1055/s-0041-1724117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Heparan sulfate proteoglycans (HSPGs) are a diverse family of polysaccharides, consisting of a core protein with glycosaminoglycan (GAG) side chains attached. The heterogeneous GAG side-chain carbohydrates consist of repeating disaccharides, with each side chain possessing a specific sulfation pattern. It is the variable sulfation pattern that allows HSPGs to interact with numerous ligands including growth factors, cytokines, chemokines, morphogens, extracellular matrix (ECM) glycoproteins, collagens, enzymes, and lipases. HSPGs are classified according to their localization within an individual cell, and include the membrane bound syndecans (SDCs) and glypicans (GPCs), with perlecan, agrin, and type-XVIII collagen secreted into the ECM. The stem cell niche is defined as the environment that circumscribes stem cells when they are in their naïve state, and includes the ECM, which provides a complex contribution to various biological processes during development and throughout life. These contributions include facilitating cell adhesion, proliferation, migration, differentiation, specification, and cell survival. In contrast, HSPGs play an anticoagulant role in thrombosis through being present on the luminal surface of cells, while also playing roles in the stimulation and inhibition of angiogenesis, highlighting their varied and systemic roles in cellular control. To fully understand the complexities of cell-cell and cell-matrix interactions, three-dimensional (3D) models such as hydrogels offer researchers exciting opportunities, such as controllable 3D in vitro environments, that more readily mimic the in vivo/in situ microenvironment. This review examines our current knowledge of HSPGs in the stem cell niche, human stem cell models, and their role in the development of 3D models that mimic the in vivo neural ECM.
Collapse
Affiliation(s)
- Ian W Peall
- Genomics Research Centre, Stem Cell and Neurogenesis Group, Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Rachel K Okolicsanyi
- Genomics Research Centre, Stem Cell and Neurogenesis Group, Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Stem Cell and Neurogenesis Group, Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Larisa M Haupt
- Genomics Research Centre, Stem Cell and Neurogenesis Group, Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| |
Collapse
|
32
|
Keil S, Gupta M, Brand M, Knopf F. Heparan sulfate proteoglycan expression in the regenerating zebrafish fin. Dev Dyn 2021; 250:1368-1380. [PMID: 33638212 DOI: 10.1002/dvdy.321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/16/2021] [Accepted: 02/10/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Heparan sulfate proteoglycan (HSPG) expression is found in many animal tissues and regulates growth factor signaling such as of Fibroblast growth factors (Fgf), Wingless/Int (Wnt) and Hedgehog (HH). Glypicans, which are GPI (glycosylphosphatidylinositol)-anchored proteins, and transmembrane-anchored syndecans represent two major HSPG protein families whose involvement in development and disease has been demonstrated. Their participation in regenerative processes both of the central nervous system and of regenerating limbs is well documented. However, whether HSPG are expressed in regenerating zebrafish fins, is currently unknown. RESULTS Here, we carried out a systematic screen of glypican and syndecan mRNA expression in regenerating zebrafish fins during the outgrowth phase. We find that 8 of the 10 zebrafish glypicans and the three known zebrafish syndecans show specific expression at 3 days post amputation. Expression is found in different domains of the regenerate, including the distal and lateral basal layers of the wound epidermis, the distal most blastema and more proximal blastema regions. CONCLUSIONS HSPG expression is prevalent in regenerating zebrafish fins. Further research is needed to delineate the function of glypican and syndecan action during zebrafish fin regeneration.
Collapse
Affiliation(s)
- Sebastian Keil
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany.,Technische Universität Dresden, Center for Healthy Aging TU Dresden, Dresden, Germany
| | - Mansi Gupta
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany.,Merus N.V, Utrecht, Netherlands
| | - Michael Brand
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Franziska Knopf
- Technische Universität Dresden, CRTD - Center for Regenerative Therapies TU Dresden, Dresden, Germany.,Technische Universität Dresden, Center for Healthy Aging TU Dresden, Dresden, Germany
| |
Collapse
|
33
|
Vieira WA, Goren S, McCusker CD. ECM-mediated positional cues are able to induce pattern, but not new positional information, during axolotl limb regeneration. PLoS One 2021; 16:e0248051. [PMID: 33667253 PMCID: PMC7935289 DOI: 10.1371/journal.pone.0248051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/18/2021] [Indexed: 11/18/2022] Open
Abstract
The Mexican Axolotl is able to regenerate missing limb structures in any position along the limb axis throughout its life and serves as an excellent model to understand the basic mechanisms of endogenous regeneration. How the new pattern of the regenerating axolotl limb is established has not been completely resolved. An accumulating body of evidence indicates that pattern formation occurs in a hierarchical fashion, which consists of two different types of positional communications. The first type (Type 1) of communication occurs between connective tissue cells, which retain memory of their original pattern information and use this memory to generate the pattern of the regenerate. The second type (Type 2) of communication occurs from connective tissue cells to other cell types in the regenerate, which don’t retain positional memory themselves and arrange themselves according to these positional cues. Previous studies suggest that molecules within the extracellular matrix (ECM) participate in pattern formation in developing and regenerating limbs. However, it is unclear whether these molecules play a role in Type 1 or Type 2 positional communications. Utilizing the Accessory Limb Model, a regenerative assay, and transcriptomic analyses in regenerates that have been reprogrammed by treatment with Retinoic Acid, our data indicates that the ECM likely facilities Type-2 positional communications during limb regeneration.
Collapse
Affiliation(s)
- Warren A. Vieira
- Department of Biology, University of Massachusetts, Boston, MA, United States of America
| | - Shira Goren
- Department of Biology, University of Massachusetts, Boston, MA, United States of America
| | - Catherine D. McCusker
- Department of Biology, University of Massachusetts, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
34
|
Akasaka-Manya K, Manya H, Nadanaka S, Kitagawa H, Kondo Y, Ishigami A, Endo T. Decreased ADAM17 expression in the lungs of α-Klotho reduced mouse. J Biochem 2021; 167:483-493. [PMID: 31951006 DOI: 10.1093/jb/mvz113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 12/11/2019] [Indexed: 01/26/2023] Open
Abstract
The deficiency of α-Klotho in mice causes phenotypes resembling human age-associated disorders at 3-4 weeks after birth and shows short lifespans of ∼2 months. One of the crucial symptoms is pulmonary emphysema, although α-Klotho is not expressed in the lungs. α-Klotho secreted from the kidneys is probably involved in the pathology of emphysema because kidney-specific knockout mice exhibit emphysematous structural changes. We examined whether any glycan changes in α-Klotho mouse lungs were observed, because α-Klotho is reported to have glycosidase activity. Here, we found the accumulation of heparan sulphate in the microsomal fraction of α-Klotho mouse lungs. Meanwhile, a disintegrin and metalloproteinase 17 (ADAM17) expression was decreased in α-Klotho mice. From these results, it is thought that the increase in heparan sulphate is due to insufficient cleavage of the core protein by ADAM17. Additionally, a reduction in α-Klotho and a decline of ADAM17 were also observed both in normal aged mice and in senescence marker protein-30 (SMP30) knockout mice, a mouse model of premature ageing. Thus, the decrease in ADAM17 is caused by the reduction in α-Klotho. These may be involved in the deterioration of lung function during ageing and may be associated with the pathology of pulmonary emphysema.
Collapse
Affiliation(s)
- Keiko Akasaka-Manya
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Hiroshi Manya
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan
| | - Satomi Nadanaka
- Laboratory of Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558, Japan
| | - Yoshitaka Kondo
- Molecular Regulation of Aging, Research Team for Functional Biogerontology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo 173-0015, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Research Team for Functional Biogerontology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo 173-0015, Japan
| | - Tamao Endo
- Molecular Glycobiology, Research Team for Mechanism of Aging, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakaecho, Itabashi-ku, Tokyo 173-0015, Japan
| |
Collapse
|
35
|
Kim GC, Cheon DH, Lee Y. Challenge to overcome current limitations of cell-penetrating peptides. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140604. [PMID: 33453413 DOI: 10.1016/j.bbapap.2021.140604] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/21/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022]
Abstract
The penetration of biological membranes is a prime obstacle for the delivery of pharmaceutical drugs. Cell-penetrating peptide (CPP) is an efficient vehicle that can deliver various cargos across the biological membranes. Since the discovery, CPPs have been rigorously studied to unveil the underlying penetrating mechanism as well as to exploit CPPs for various biomedical applications. This review will focus on the various strategies to overcome current limitations regarding stability, selectivity, and efficacy of CPPs.
Collapse
Affiliation(s)
- Gyu Chan Kim
- Department of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea
| | - Dae Hee Cheon
- Department of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea
| | - Yan Lee
- Department of Chemistry, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
36
|
Alshehri MA, Alshehri MM, Albalawi NN, Al-Ghamdi MA, Al-Gayyar MMH. Heparan sulfate proteoglycans and their modification as promising anticancer targets in hepatocellular carcinoma. Oncol Lett 2021; 21:173. [PMID: 33552290 PMCID: PMC7798035 DOI: 10.3892/ol.2021.12434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/08/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common types of primary liver cancer. Despite advancements in the treatment strategies of HCC, there is an urgent requirement to identify and develop novel therapeutic drugs that do not lead to resistance. These novel agents should have the potential to influence the primary mechanisms participating in the pathogenesis of HCC. Heparan sulfate proteoglycans (HSPGs) are major elements of the extracellular matrix that perform structural and signaling functions. HSPGs protect against invasion of tumor cells by preventing cell infiltration and intercellular adhesion. Several enzymes, such as heparanase, matrix metalloproteinase-9 and sulfatase-2, have been reported to affect HSPGs, leading to their degradation and thus enhancing tumor invasion. In addition, some compounds that are produced from the degradation of HSPGs, including glypican-3 and syndecan-1, enhance tumor progression. Thus, the identification of enzymes that affect HSPGs or their degradation products in HCC may lead to the development of novel therapeutic targets. The present review discusses the main enzymes and compounds associated with HSPGs, and their involvement with the pathogenicity of HCC.
Collapse
Affiliation(s)
- Mohammed A Alshehri
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Moath M Alshehri
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Naif N Albalawi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Moshari A Al-Ghamdi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Mohammed M H Al-Gayyar
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia.,Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.,Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia
| |
Collapse
|
37
|
Filipek-Górniok B, Habicher J, Ledin J, Kjellén L. Heparan Sulfate Biosynthesis in Zebrafish. J Histochem Cytochem 2020; 69:49-60. [PMID: 33216642 DOI: 10.1369/0022155420973980] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The biosynthesis of heparan sulfate (HS) proteoglycans occurs in the Golgi compartment of cells and will determine the sulfation pattern of HS chains, which in turn will have a large impact on the biological activity of the proteoglycans. Earlier studies in mice have demonstrated the importance of HS for embryonic development. In this review, the enzymes participating in zebrafish HS biosynthesis, along with a description of enzyme mutants available for functional studies, are presented. The consequences of the zebrafish genome duplication and maternal transcript contribution are briefly discussed as are the possibilities of CRISPR/Cas9 methodologies to use the zebrafish model system for studies of biosynthesis as well as proteoglycan biology.
Collapse
Affiliation(s)
- Beata Filipek-Górniok
- Department of Organismal Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Judith Habicher
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Johan Ledin
- Department of Organismal Biology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lena Kjellén
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
38
|
Ravikumar M, Smith RAA, Nurcombe V, Cool SM. Heparan Sulfate Proteoglycans: Key Mediators of Stem Cell Function. Front Cell Dev Biol 2020; 8:581213. [PMID: 33330458 PMCID: PMC7710810 DOI: 10.3389/fcell.2020.581213] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are an evolutionarily ancient subclass of glycoproteins with exquisite structural complexity. They are ubiquitously expressed across tissues and have been found to exert a multitude of effects on cell behavior and the surrounding microenvironment. Evidence has shown that heterogeneity in HSPG composition is crucial to its functions as an essential scaffolding component in the extracellular matrix as well as a vital cell surface signaling co-receptor. Here, we provide an overview of the significance of HSPGs as essential regulators of stem cell function. We discuss the various roles of HSPGs in distinct stem cell types during key physiological events, from development through to tissue homeostasis and regeneration. The contribution of aberrant HSPG production to altered stem cell properties and dysregulated cellular homeostasis characteristic of cancer is also reviewed. Finally, we consider approaches to better understand and exploit the multifaceted functions of HSPGs in influencing stem cell characteristics for cell therapy and associated culture expansion strategies.
Collapse
Affiliation(s)
- Maanasa Ravikumar
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Raymond Alexander Alfred Smith
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Victor Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College London, Singapore, Singapore
| | - Simon M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
39
|
Egorova A, Bogner E, Novoselova E, Zorn KM, Ekins S, Makarov V. Dispirotripiperazine-core compounds, their biological activity with a focus on broad antiviral property, and perspectives in drug design (mini-review). Eur J Med Chem 2020; 211:113014. [PMID: 33218683 PMCID: PMC7658596 DOI: 10.1016/j.ejmech.2020.113014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/26/2020] [Accepted: 11/08/2020] [Indexed: 12/31/2022]
Abstract
Viruses are obligate intracellular parasites and have evolved to enter the host cell. To gain access they come into contact with the host cell through an initial adhesion, and some viruses from different genus may use heparan sulfate proteoglycans for it. The successful inhibition of this early event of the infection by synthetic molecules has always been an attractive target for medicinal chemists. Numerous reports have yielded insights into the function of compounds based on the dispirotripiperazine scaffold. Analysis suggests that this is a structural requirement for inhibiting the interactions between viruses and cell-surface heparan sulfate proteoglycans, thus preventing virus entry and replication. This review summarizes our current knowledge about the early history of development, synthesis, structure-activity relationships and antiviral evaluation of dispirotripiperazine-based compounds and where they are going in the future.
Collapse
Affiliation(s)
- Anna Egorova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow, Russia
| | - Elke Bogner
- Institute of Virology, Charité Universitätsmedizin Berlin, Charité Campus Mitte, Chariteplatz 1, 10117, Berlin, Germany
| | - Elena Novoselova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow, Russia
| | - Kimberley M Zorn
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab, 3510, Raleigh, NC, USA
| | - Vadim Makarov
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, 119071, Moscow, Russia.
| |
Collapse
|
40
|
Benetó N, Vilageliu L, Grinberg D, Canals I. Sanfilippo Syndrome: Molecular Basis, Disease Models and Therapeutic Approaches. Int J Mol Sci 2020; 21:E7819. [PMID: 33105639 PMCID: PMC7659972 DOI: 10.3390/ijms21217819] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022] Open
Abstract
Sanfilippo syndrome or mucopolysaccharidosis III is a lysosomal storage disorder caused by mutations in genes responsible for the degradation of heparan sulfate, a glycosaminoglycan located in the extracellular membrane. Undegraded heparan sulfate molecules accumulate within lysosomes leading to cellular dysfunction and pathology in several organs, with severe central nervous system degeneration as the main phenotypical feature. The exact molecular and cellular mechanisms by which impaired degradation and storage lead to cellular dysfunction and neuronal degeneration are still not fully understood. Here, we compile the knowledge on this issue and review all available animal and cellular models that can be used to contribute to increase our understanding of Sanfilippo syndrome disease mechanisms. Moreover, we provide an update in advances regarding the different and most successful therapeutic approaches that are currently under study to treat Sanfilippo syndrome patients and discuss the potential of new tools such as induced pluripotent stem cells to be used for disease modeling and therapy development.
Collapse
Affiliation(s)
- Noelia Benetó
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, CIBERER, IBUB, IRSJD, E-08028 Barcelona, Spain; (N.B.); (L.V.); (D.G.)
| | - Lluïsa Vilageliu
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, CIBERER, IBUB, IRSJD, E-08028 Barcelona, Spain; (N.B.); (L.V.); (D.G.)
| | - Daniel Grinberg
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, CIBERER, IBUB, IRSJD, E-08028 Barcelona, Spain; (N.B.); (L.V.); (D.G.)
| | - Isaac Canals
- Stem Cells, Aging and Neurodegeneration Group, Department of Clinical Sciences, Neurology, Lund Stem Cell Center, Lund University, SE-22184 Lund, Sweden
| |
Collapse
|
41
|
Balomenos P, Dragomir A, Tsakalidis AK, Bezerianos A. Identification of differentially expressed subpathways via a bilevel consensus scoring of network topology and gene expression. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2020; 2020:5316-5319. [PMID: 33019184 DOI: 10.1109/embc44109.2020.9176556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Identifying differentially expressed subpathways connected to the emergence of a disease that can be considered as candidates for pharmacological intervention, with minimal off-target effects, is a daunting task. In this direction, we present a bilevel subpathway analysis method to identify differentially expressed subpathways that are connected with an experimental condition, while taking into account potential crosstalks between subpathways which arise due to their connectivity in a combined multi-pathway network. The efficacy of the method is demonstrated on a hematopoietic stem cell aging dataset, with findings corroborated using recent literature.
Collapse
|
42
|
Griffin ME, Sorum AW, Miller GM, Goddard WA, Hsieh-Wilson LC. Sulfated glycans engage the Ang-Tie pathway to regulate vascular development. Nat Chem Biol 2020; 17:178-186. [PMID: 33020664 PMCID: PMC8087285 DOI: 10.1038/s41589-020-00657-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 08/20/2020] [Indexed: 12/13/2022]
Abstract
The angiopoietin (Ang)/Tie pathway is essential for the proper maturation and remodeling of the vasculature. Despite its importance in disease, the mechanisms that control signal transduction through this pathway are poorly understood. Here, we demonstrate that heparan sulfate glycosaminoglycans (HS GAGs) regulate Ang/Tie signaling through direct interactions with both Ang ligands and the Tie1 receptor. HS GAGs bound to Ang1/4 ligands and formed ternary Ang-Tie2 receptor complexes, thereby potentiating endothelial survival signaling. In addition, we found that HS GAGs are novel ligands for the orphan receptor Tie1. The HS-Tie1 interaction promoted Tie1-Tie2 heterodimerization and enhanced Tie1 stability within the mature vasculature. Loss of HS-Tie1 binding using CRISPR/Cas9-mediated mutagenesis in vivo led to decreased Tie protein levels, pathway suppression, and aberrant retinal vascularization. Together, these results reveal that sulfated glycans use dual mechanisms to regulate Ang/Tie signaling and are important for the development and maintenance of the vasculature.
Collapse
Affiliation(s)
- Matthew E Griffin
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Alexander W Sorum
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Gregory M Miller
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - William A Goddard
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA.,Materials and Process Simulation Center and Joint Center for Artificial Photosynthesis, California Institute of Technology, Pasadena, CA, USA
| | - Linda C Hsieh-Wilson
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
43
|
Lan Y, Liu Y, He Y, Liu F, Xv H, Feng K, Zhang Z, Shi Z, Zhang X, Zhang L. A single injection of bleomycin reduces glycosaminoglycan sulfation up to 30 days in the C57BL/6 mouse model of lung fibrosis. Int J Biol Macromol 2020; 160:319-327. [PMID: 32422263 DOI: 10.1016/j.ijbiomac.2020.05.087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 04/06/2020] [Accepted: 05/10/2020] [Indexed: 12/13/2022]
Abstract
Bleomycin is a clinically used anticancer drug, but it induces lung fibrosis in certain cancer patients with unknown mechanism. Glycosaminoglycans (GAGs) are required for lung morphogenesis during animal development. In current study, GAG disaccharides including heparan sulfate (HS) and chondroitin sulfate (CS) from bleomycin-induced and control lung tissues in lung fibrosis mouse model were tagged with 1-phenyl-3-methyl-5-pyrazolone (PMP) and deuterated PMP, respectively. The differentially isotope-tagged disaccharides were quantitatively compared by LC-MS. At day 10, the amount of CS disaccharides (U0a0, U0a6, and U0a4) and non-sulfated HS disaccharide (U0A0) were increased by 1.3-, 1.6-, 11.7-, and 2.2-fold, respectively, whereas the amount of CS disaccharide (U0a2), hyaluranan disaccharide (UβA0), and six HS disaccharides (U0A6, U2A0, U0H6, U0S0, U2S0, and U2S6) were decreased from1.1- to 14.3-fold compared to that of the controls. At day 15, under-sulfation of both HS and CS disaccharides was persisted. At day 30, the CS disaccharide compositions were recovered to that of the control levels whereas the HS were still remarkably under-sulfated. In conclusion, GAGs, especially HS, from fibrotic lungs induced by a single injection of bleomycin were significantly under-sulfated up to 30 days, suggesting GAGs might be another class of defective signaling molecules involved in bleomycin-induced lung fibrosis.
Collapse
Affiliation(s)
- Ying Lan
- Systems Biology and Medicine Center for Complex Diseases, the Affiliated Hospital of Qingdao University, Qingdao 266003, China; College of Food Science and engineering, Northwest A&F University, 712100, China
| | - Yong Liu
- Systems Biology and Medicine Center for Complex Diseases, the Affiliated Hospital of Qingdao University, Qingdao 266003, China; School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Yanli He
- Systems Biology and Medicine Center for Complex Diseases, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Feng Liu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Weifang Medical College, Weifang 261031, China
| | - Huixin Xv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Kun Feng
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Zhenkun Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Zhaoyu Shi
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Xiaolei Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Lijuan Zhang
- Systems Biology and Medicine Center for Complex Diseases, the Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
44
|
Puri S, Coulson-Thomas YM, Gesteira TF, Coulson-Thomas VJ. Distribution and Function of Glycosaminoglycans and Proteoglycans in the Development, Homeostasis and Pathology of the Ocular Surface. Front Cell Dev Biol 2020; 8:731. [PMID: 32903857 PMCID: PMC7438910 DOI: 10.3389/fcell.2020.00731] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/15/2020] [Indexed: 12/20/2022] Open
Abstract
The ocular surface, which forms the interface between the eye and the external environment, includes the cornea, corneoscleral limbus, the conjunctiva and the accessory glands that produce the tear film. Glycosaminoglycans (GAGs) and proteoglycans (PGs) have been shown to play important roles in the development, hemostasis and pathology of the ocular surface. Herein we review the current literature related to the distribution and function of GAGs and PGs within the ocular surface, with focus on the cornea. The unique organization of ECM components within the cornea is essential for the maintenance of corneal transparency and function. Many studies have described the importance of GAGs within the epithelial and stromal compartment, while very few studies have analyzed the ECM of the endothelial layer. Importantly, GAGs have been shown to be essential for maintaining corneal homeostasis, epithelial cell differentiation and wound healing, and, more recently, a role has been suggested for the ECM in regulating limbal stem cells, corneal innervation, corneal inflammation, corneal angiogenesis and lymphangiogenesis. Reports have also associated genetic defects of the ECM to corneal pathologies. Thus, we also highlight the role of different GAGs and PGs in ocular surface homeostasis, as well as in pathology.
Collapse
Affiliation(s)
- Sudan Puri
- College of Optometry, University of Houston, Houston, TX, United States
| | - Yvette M Coulson-Thomas
- Molecular Biology Section, Department of Biochemistry, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Tarsis F Gesteira
- College of Optometry, University of Houston, Houston, TX, United States.,Optimvia, LLC, Batavia, OH, United States
| | | |
Collapse
|
45
|
Vallet SD, Clerc O, Ricard-Blum S. Glycosaminoglycan-Protein Interactions: The First Draft of the Glycosaminoglycan Interactome. J Histochem Cytochem 2020; 69:93-104. [PMID: 32757871 DOI: 10.1369/0022155420946403] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The six mammalian glycosaminoglycans (GAGs), chondroitin sulfate, dermatan sulfate, heparin, heparan sulfate, hyaluronan, and keratan sulfate, are linear polysaccharides. Except for hyaluronan, they are sulfated to various extent, and covalently attached to proteins to form proteoglycans. GAGs interact with growth factors, morphogens, chemokines, extracellular matrix proteins and their bioactive fragments, receptors, lipoproteins, and pathogens. These interactions mediate their functions, from embryonic development to extracellular matrix assembly and regulation of cell signaling in various physiological and pathological contexts such as angiogenesis, cancer, neurodegenerative diseases, and infections. We give an overview of GAG-protein interactions (i.e., specificity and chemical features of GAG- and protein-binding sequences), and review the available GAG-protein interaction networks. We also provide the first comprehensive draft of the GAG interactome composed of 832 biomolecules (827 proteins and five GAGs) and 932 protein-GAG interactions. This network is a scaffold, which in the future should integrate structures of GAG-protein complexes, quantitative data of the abundance of GAGs in tissues to build tissue-specific interactomes, and GAG interactions with metal ions such as calcium, which plays a major role in the assembly of the extracellular matrix and its interactions with cells. This contextualized interactome will be useful to identify druggable GAG-protein interactions for therapeutic purpose.
Collapse
Affiliation(s)
- Sylvain D Vallet
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, Villeurbanne Cedex, France
| | - Olivier Clerc
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, Villeurbanne Cedex, France
| | - Sylvie Ricard-Blum
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, Villeurbanne Cedex, France
| |
Collapse
|
46
|
The conformation of the idopyranose ring revisited: How subtle O-substituent induced changes can be deduced from vicinal 1H-NMR coupling constants. Carbohydr Res 2020; 496:108052. [PMID: 32738719 DOI: 10.1016/j.carres.2020.108052] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/17/2020] [Accepted: 05/27/2020] [Indexed: 01/18/2023]
Abstract
The idopyranose ring plays a pivotal role in the conformational, dynamical, and intermolecular binding aspects of glycosaminoglycans like heparin and dermatan sulfate and it was early on assigned a role in the Sugar Code governing biological recognition processes. There is consensus that next to the two canonical 1C4 and 4C1 chair conformations, the conformational space accessible to the idopyranose ring entails a 2SO skew-boat conformation, but the equilibrium between these three ring puckers has evaded satisfactory quantification. In this study a meta-analysis of X-ray solid-state data and vicinal NMR coupling constants is presented, based on the Truncated Fourier Puckering (TFP) formalism and the generalized Karplus (CAGPLUS) equation. This approach yields a model-free, granular and consistent reckoning of 159 idopyranose solution puckering equilibria studied by NMR and allows us to reproduce the involved 636 NMR vicinal couplings with an overall residual RMS(Jobs-Jcalc) of 0.184 Hz. Our analyses show that for all ring systems examined, the idopyranosyl chair conformations take up the same ring pucker irrespective of the ring substituent pattern or a vast variety in experimental conditions. Instead, it is the (skew-)boat conformation that adapts to the substitution pattern of the idopyranose ring or a specific sulfation pattern of neighboring saccharides. All idopyranose rings are involved in conformational equilibria that subsume the aforementioned conformers which turn out to differ only a few kJ/mole in conformational energy. Thus, the plasticity and flexibility of idopyranose remains intact under practically all circumstances and, as the glycosidic linkages in heparin are considered to be relatively stiff, the iduronic moiety functions as the linchpin of heparin flexibility thereby being rather a "space(r)" than a "letter" in the alleged Sugar Code alphabet.
Collapse
|
47
|
Cui H, Cheng X, Batool T, Zhang X, Li JP. Glucuronyl C5-epimerase is crucial for epithelial cell maturation during embryonic lung development. Glycobiology 2020; 31:223-230. [PMID: 32651954 DOI: 10.1093/glycob/cwaa065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/28/2020] [Accepted: 07/06/2020] [Indexed: 12/23/2022] Open
Abstract
Glucuronyl C5-epimerase (Hsepi) is a key enzyme in the biosynthesis of heparan sulfate that is a sulfated polysaccharide expressed on the cell surface and in the extracellular matrix of alveolar walls and blood vessels. Targeted interruption of the Hsepi gene, Glce, in mice resulted in neonatal lethality, which is most likely due to lung atelectasis. In this study, we examined the potential mechanisms behind the defect in lung development. Histological analysis of the lungs from embryos revealed no difference in the morphology between wild-type and mutant animals up to E16.5. This suggests that the initial events leading to formation of the lung primordium and branching morphogenesis are not disturbed. However, the distal lung of E17.5-18.5 mutants is still populated by epithelial tubules, lacking the typical saccular structural characteristic of a normal E17.5 lung. Immunostaining revealed strong signals of surfactant protein-C, but a weaker signal of T1α in the mutant lungs in comparison to WT littermates, suggesting differentiation of type I alveolar epithelial cells (AT1) is impaired. One of the parameters contributed to the failure of AT1 maturation is reduced vascularization in the developing lungs.
Collapse
Affiliation(s)
- Hao Cui
- College of Life Science, Jiangxi Normal University, 99 Ziyang Avenue, 330022, Nanchang, China.,Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Box 582, Husargatan 3, S-75123, Uppsala, Sweden
| | - Xiaowen Cheng
- Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Box 582, Husargatan 3, S-75123, Uppsala, Sweden
| | - Tahira Batool
- Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Box 582, Husargatan 3, S-75123, Uppsala, Sweden
| | - Xiao Zhang
- Department of Neuroscience and Pharmacology, University of Uppsala, Box 582, Husargatan 3, S-75123, Uppsala, Sweden
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, SciLifeLab Uppsala, The Biomedical Center, University of Uppsala, Box 582, Husargatan 3, S-75123, Uppsala, Sweden
| |
Collapse
|
48
|
Gulberti S, Mao X, Bui C, Fournel-Gigleux S. The role of heparan sulfate maturation in cancer: A focus on the 3O-sulfation and the enigmatic 3O-sulfotransferases (HS3STs). Semin Cancer Biol 2020; 62:68-85. [DOI: 10.1016/j.semcancer.2019.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 01/05/2023]
|
49
|
Lindsay SL, McCanney GA, Willison AG, Barnett SC. Multi-target approaches to CNS repair: olfactory mucosa-derived cells and heparan sulfates. Nat Rev Neurol 2020; 16:229-240. [PMID: 32099190 DOI: 10.1038/s41582-020-0311-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) remains one of the biggest challenges in the development of neuroregenerative therapeutics. Cell transplantation is one of numerous experimental strategies that have been identified and tested for efficacy at both preclinical and clinical levels in recent years. In this Review, we briefly discuss the state of human olfactory cell transplantation as a therapy, considering both its current clinical status and its limitations. Furthermore, we introduce a mesenchymal stromal cell derived from human olfactory tissue, which has the potential to induce multifaceted reparative effects in the environment within and surrounding the lesion. We argue that no single therapy will be sufficient to treat SCI effectively and that a combination of cell-based, rehabilitation and pharmaceutical interventions is the most promising approach to aid repair. For this reason, we also introduce a novel pharmaceutical strategy based on modifying the activity of heparan sulfate, an important regulator of a wide range of biological cell functions. The multi-target approach that is exemplified by these types of strategies will probably be necessary to optimize SCI treatment.
Collapse
Affiliation(s)
- Susan L Lindsay
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - George A McCanney
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Alice G Willison
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Susan C Barnett
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
50
|
Rutledge EA, McMahon AP. Mutational analysis of genes with ureteric progenitor cell-specific expression in branching morphogenesis of the mouse kidney. Dev Dyn 2020; 249:765-774. [PMID: 32017326 DOI: 10.1002/dvdy.157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/09/2020] [Accepted: 01/27/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Ureteric progenitor cells (UPCs) within the branch tips of the arborizing ureteric epithelium of the kidney's developing collecting system establish the shape and cellular organization of the collecting network, and drive the nephrogenic program through their interactions with nephron progenitor cells. In a previous study, expression screening identified a cohort of genes showing UPC-enriched expression including D17H6S56E-5, Hs3st3a1, Hs3st3b1, and Tmem59l. Each of these is also enriched in branch tips of assembling airways of the developing lungs. Here, we used Crispr-CAS9 directed gene editing to mutate each of these targets to address their potential role(s) in UPC programs. RESULTS Single (D17H6S56E-5 and Tmem59l) and double (Hs3st3a1 and Hs3st3b1) mutants were viable, fertile, and displayed varying frequencies of ureter duplications and no overt lung phenotype. Ureter duplications arise spontaneously through multiple outgrowths of the ureteric bud at the onset of kidney development. Tmem59l mutants and Hs3st3a1/Hs3st3b1 compound mutants showed a weakly penetrant, but statistically significant increase in duplicated ureters compared to C57BL6/J and SW wild-type mouse strains. CONCLUSIONS Tmem59l and Hs3st3a1/Hs3st3b1 activities contribute to the regulatory programs restricting ureteric outgrowth in the developing mouse kidney. However, the low penetrance of the observed phenotype precludes a detailed analysis of their specific actions.
Collapse
Affiliation(s)
- Elisabeth A Rutledge
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|