1
|
Wu Z, Wang Y, Liu W, Lu M, Shi J. The role of neuropilin in bone/cartilage diseases. Life Sci 2024; 346:122630. [PMID: 38614296 DOI: 10.1016/j.lfs.2024.122630] [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: 12/10/2023] [Revised: 03/12/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Bone remodeling is the balance between osteoblasts and osteoclasts. Bone diseases such as osteoporosis and osteoarthritis are associated with imbalanced bone remodeling. Skeletal injury leads to limited motor function and pain. Neurophilin was initially identified in axons, and its various ligands and roles in bone remodeling, angiogenesis, neuropathic pain and immune regulation were later discovered. Neurophilin promotes osteoblast mineralization and inhibits osteoclast differentiation and its function. Neuropolin-1 provides channels for immune cell chemotaxis and cytokine diffusion and leads to pain. Neuropolin-1 regulates the proportion of T helper type 17 (Th17) and regulatory T cells (Treg cells), and affects bone immunity. Vascular endothelial growth factors (VEGF) combine with neuropilin and promote angiogenesis. Class 3 semaphorins (Sema3a) compete with VEGF to bind neuropilin, which reduces angiogenesis and rejects sympathetic nerves. This review elaborates on the structure and general physiological functions of neuropilin and summarizes the role of neuropilin and its ligands in bone and cartilage diseases. Finally, treatment strategies and future research directions based on neuropilin are proposed.
Collapse
Affiliation(s)
- Zuping Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310016, China
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310016, China
| | - Wei Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310016, China
| | - Mingcheng Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310016, China
| | - Jiejun Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310016, China.
| |
Collapse
|
2
|
Hunter C, Derksen T, Makhsous S, Doll M, Perez SR, Scott NE, Willis LM. Site-specific immobilization of the endosialidase reveals QSOX2 is a novel polysialylated protein. Glycobiology 2024; 34:cwae026. [PMID: 38489772 PMCID: PMC11031136 DOI: 10.1093/glycob/cwae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 03/04/2024] [Accepted: 03/13/2024] [Indexed: 03/17/2024] Open
Abstract
Polysialic acid (polySia) is a linear polymer of α2,8-linked sialic acid residues that is of fundamental biological interest due to its pivotal roles in the regulation of the nervous, immune, and reproductive systems in healthy human adults. PolySia is also dysregulated in several chronic diseases, including cancers and mental health disorders. However, the mechanisms underpinning polySia biology in health and disease remain largely unknown. The polySia-specific hydrolase, endoneuraminidase NF (EndoN), and the catalytically inactive polySia lectin EndoNDM, have been extensively used for studying polySia. However, EndoN is heat stable and remains associated with cells after washing. When studying polySia in systems with multiple polysialylated species, the residual EndoN that cannot be removed confounds data interpretation. We developed a strategy for site-specific immobilization of EndoN on streptavidin-coated magnetic beads. We showed that immobilizing EndoN allows for effective removal of the enzyme from samples, while retaining hydrolase activity. We used the same strategy to immobilize the polySia lectin EndoNDM, which enabled the enrichment of polysialylated proteins from complex mixtures such as serum for their identification via mass spectrometry. We used this methodology to identify a novel polysialylated protein, QSOX2, which is secreted from the breast cancer cell line MCF-7. This method of site-specific immobilization can be utilized for other enzymes and lectins to yield insight into glycobiology.
Collapse
Affiliation(s)
- Carmanah Hunter
- Department of Biological Sciences, University of Alberta, 116 St & 85 Ave, Edmonton, AB, T6G 2R3, Canada
| | - Tahlia Derksen
- Department of Biological Sciences, University of Alberta, 116 St & 85 Ave, Edmonton, AB, T6G 2R3, Canada
| | - Sogand Makhsous
- Department of Biological Sciences, University of Alberta, 116 St & 85 Ave, Edmonton, AB, T6G 2R3, Canada
| | - Matt Doll
- Department of Biological Sciences, University of Alberta, 116 St & 85 Ave, Edmonton, AB, T6G 2R3, Canada
| | - Samantha Rodriguez Perez
- Department of Biological Sciences, University of Alberta, 116 St & 85 Ave, Edmonton, AB, T6G 2R3, Canada
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Lisa M Willis
- Department of Biological Sciences, University of Alberta, 116 St & 85 Ave, Edmonton, AB, T6G 2R3, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, 116 St & 85 Ave, Edmonton, AB, T6G 2R3, Canada
| |
Collapse
|
3
|
Dhupar R, Powers AA, Eisenberg SH, Gemmill RM, Bardawil CE, Udoh HM, Cubitt A, Nangle LA, Soloff AC. Orchestrating Resilience: How Neuropilin-2 and Macrophages Contribute to Cardiothoracic Disease. J Clin Med 2024; 13:1446. [PMID: 38592275 PMCID: PMC10934188 DOI: 10.3390/jcm13051446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 04/10/2024] Open
Abstract
Immunity has evolved to balance the destructive nature of inflammation with wound healing to overcome trauma, infection, environmental insults, and rogue malignant cells. The inflammatory response is marked by overlapping phases of initiation, resolution, and post-resolution remodeling. However, the disruption of these events can lead to prolonged tissue damage and organ dysfunction, resulting long-term disease states. Macrophages are the archetypic phagocytes present within all tissues and are important contributors to these processes. Pleiotropic and highly plastic in their responses, macrophages support tissue homeostasis, repair, and regeneration, all while balancing immunologic self-tolerance with the clearance of noxious stimuli, pathogens, and malignant threats. Neuropilin-2 (Nrp2), a promiscuous co-receptor for growth factors, semaphorins, and integrins, has increasingly been recognized for its unique role in tissue homeostasis and immune regulation. Notably, recent studies have begun to elucidate the role of Nrp2 in both non-hematopoietic cells and macrophages with cardiothoracic disease. Herein, we describe the unique role of Nrp2 in diseases of the heart and lung, with an emphasis on Nrp2 in macrophages, and explore the potential to target Nrp2 as a therapeutic intervention.
Collapse
Affiliation(s)
- Rajeev Dhupar
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Surgical and Research Services, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Amy A. Powers
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Seth H. Eisenberg
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Robert M. Gemmill
- Division of Hematology/Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Charles E. Bardawil
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Hannah M. Udoh
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
| | - Andrea Cubitt
- aTyr Pharma, San Diego, CA 92121, USA; (A.C.); (L.A.N.)
| | | | - Adam C. Soloff
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (R.D.); (H.M.U.)
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Surgical and Research Services, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| |
Collapse
|
4
|
Kannan S, Rutkowski JM. VEGFR-3 signaling in macrophages: friend or foe in disease? Front Immunol 2024; 15:1349500. [PMID: 38464522 PMCID: PMC10921555 DOI: 10.3389/fimmu.2024.1349500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/01/2024] [Indexed: 03/12/2024] Open
Abstract
Lymphatic vessels have been increasingly appreciated in the context of immunology not only as passive conduits for immune and cancer cell transport but also as key in local tissue immunomodulation. Targeting lymphatic vessel growth and potential immune regulation often takes advantage of vascular endothelial growth factor receptor-3 (VEGFR-3) signaling to manipulate lymphatic biology. A receptor tyrosine kinase, VEGFR-3, is highly expressed on lymphatic endothelial cells, and its signaling is key in lymphatic growth, development, and survival and, as a result, often considered to be "lymphatic-specific" in adults. A subset of immune cells, notably of the monocyte-derived lineage, have been identified to express VEGFR-3 in tissues from the lung to the gut and in conditions as varied as cancer and chronic kidney disease. These VEGFR-3+ macrophages are highly chemotactic toward the VEGFR-3 ligands VEGF-C and VEGF-D. VEGFR-3 signaling has also been implicated in dictating the plasticity of these cells from pro-inflammatory to anti-inflammatory phenotypes. Conversely, expression may potentially be transient during monocyte differentiation with unknown effects. Macrophages play critically important and varied roles in the onset and resolution of inflammation, tissue remodeling, and vasculogenesis: targeting lymphatic vessel growth and immunomodulation by manipulating VEGFR-3 signaling may thus impact macrophage biology and their impact on disease pathogenesis. This mini review highlights the studies and pathologies in which VEGFR-3+ macrophages have been specifically identified, as well as the activity and polarization changes that macrophage VEGFR-3 signaling may elicit, and affords some conclusions as to the importance of macrophage VEGFR-3 signaling in disease.
Collapse
Affiliation(s)
| | - Joseph M. Rutkowski
- Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX, United States
| |
Collapse
|
5
|
Hatanaka R, Hane M, Hayakawa K, Morishita S, Ohno S, Yamaguchi Y, Wu D, Kitajima K, Sato C. Identification of a buried β-strand as a novel disease-related motif in the human polysialyltransferases. J Biol Chem 2024; 300:105564. [PMID: 38103644 PMCID: PMC10828065 DOI: 10.1016/j.jbc.2023.105564] [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: 07/16/2023] [Revised: 11/26/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023] Open
Abstract
The polysialyltransferases ST8SIA2 and ST8SIA4 and their product, polysialic acid (polySia), are known to be related to cancers and mental disorders. ST8SIA2 and ST8SIA4 have conserved amino acid (AA) sequence motifs essential for the synthesis of the polySia structures on the neural cell adhesion molecule. To search for a new motif in the polysialyltransferases, we adopted the in silico Individual Meta Random Forest program that can predict disease-related AA substitutions. The Individual Meta Random Forest program predicted a new eight-amino-acids sequence motif consisting of highly pathogenic AA residues, thus designated as the pathogenic (P) motif. A series of alanine point mutation experiments in the pathogenic motif (P motif) showed that most P motif mutants lost the polysialylation activity without changing the proper enzyme expression levels or localization in the Golgi. In addition, we evaluated the enzyme stability of the P motif mutants using newly established calculations of mutation energy, demonstrating that the subtle change of the conformational energy regulates the activity. In the AlphaFold2 model, we found that the P motif was a buried β-strand underneath the known surface motifs unique to ST8SIA2 and ST8SIA4. Taken together, the P motif is a novel buried β-strand that regulates the full activity of polysialyltransferases from the inside of the molecule.
Collapse
Affiliation(s)
- Rina Hatanaka
- Integrated Glyco-BioMedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan; Bioscience and Biotechnology Center, Nagoya University, Nagoya, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Masaya Hane
- Integrated Glyco-BioMedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan; Bioscience and Biotechnology Center, Nagoya University, Nagoya, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Kaito Hayakawa
- Integrated Glyco-BioMedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan; Bioscience and Biotechnology Center, Nagoya University, Nagoya, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Sayo Morishita
- Integrated Glyco-BioMedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan; Bioscience and Biotechnology Center, Nagoya University, Nagoya, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Shiho Ohno
- Division of Structural Biology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yoshiki Yamaguchi
- Division of Structural Biology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Di Wu
- Integrated Glyco-BioMedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan; Bioscience and Biotechnology Center, Nagoya University, Nagoya, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Ken Kitajima
- Integrated Glyco-BioMedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan; Bioscience and Biotechnology Center, Nagoya University, Nagoya, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Chihiro Sato
- Integrated Glyco-BioMedical Research Center (iGMED), Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan; Bioscience and Biotechnology Center, Nagoya University, Nagoya, Japan; Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan.
| |
Collapse
|
6
|
Qiao X, Liu C, Wang W, Yang C, Li M, Yi Q, Kong N, Qiu L, Liu X, Wang L, Song L. A neural cell adhesion molecule from oyster Crassostrea gigas: Molecular identification and immune functional characterization. Int J Biol Macromol 2023; 247:125756. [PMID: 37429340 DOI: 10.1016/j.ijbiomac.2023.125756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/02/2023] [Accepted: 07/07/2023] [Indexed: 07/12/2023]
Abstract
Neural cell adhesion molecules (NCAMs) are large cell-surface glycoproteins playing important roles in cell-cell and cell-extracellular matrix interactions in nervous system. Recent study identified a homologue of NCAM (CgNCAM) from the Pacific oyster Crassostrea gigas. Its ORF was of 2634 bp which encodes a protein (877 amino acids) consisting of five immunoglobulin domains and two fibronectin type III domains. CgNCAM transcripts were broadly distributed in oyster tissues especially in mantle, labial palp and haemolymph. CgNCAM showed up-regulated expression in haemocytes of oysters after Vibrio splendidus and Staphylococcus aureus stimulation. The recombinant CgNCAM protein (rCgNCAM) was able to bind manose, lipopolysaccharide and glucan, as well as different microbes including Gram-negative bacteria and fungi. rCgNCAM displayed bacterial agglutination and hemagglutination activity. CgNCAM improved the phagocytosis of haemocytes towards V. splendidus by regulating the expression of CgIntegrin, CgRho J and CgMAPKK. Moreover, CgNCAM was involved in the extracellular trap establishment of haemocytes after V. splendidus stimulation. The results collectively indicated that CgNCAM acted as a recognition receptor executing multiple immune functions to recognize and eliminate invading microorganisms in innate immunity of oysters.
Collapse
Affiliation(s)
- Xue Qiao
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Conghui Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Weilin Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Chuanyan Yang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Meijia Li
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Qilin Yi
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Ning Kong
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Limei Qiu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Xiyang Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China.
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian 116023, China; Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian 116023, China.
| |
Collapse
|
7
|
Shinde P, Kiepas A, Zhang L, Sudhir S, Konstantopoulos K, Stamatos NM. Polysialylation controls immune function of myeloid cells in murine model of pneumococcal pneumonia. Cell Rep 2023; 42:112648. [PMID: 37339052 PMCID: PMC10592499 DOI: 10.1016/j.celrep.2023.112648] [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: 11/01/2022] [Revised: 04/13/2023] [Accepted: 05/31/2023] [Indexed: 06/22/2023] Open
Abstract
Polysialic acid (polySia) is a post-translational modification of a select group of cell-surface proteins that guides cellular interactions. As the overall impact of changes in expression of this glycan on leukocytes during infection is not known, we evaluate the immune response of polySia-deficient ST8SiaIV-/- mice infected with Streptococcus pneumoniae (Spn). Compared with wild-type (WT) mice, ST8SiaIV-/- mice are less susceptible to infection and clear Spn from airways faster, with alveolar macrophages demonstrating greater viability and phagocytic activity. Leukocyte pulmonary recruitment, paradoxically, is diminished in infected ST8SiaIV-/- mice, corroborated by adoptive cell transfer, microfluidic migration experiments, and intravital microscopy, and possibly explained by dysregulated ERK1/2 signaling. PolySia is progressively lost from neutrophils and monocytes migrating from bone marrow to alveoli in Spn-infected WT mice, consistent with changing cellular functions. These data highlight multidimensional effects of polySia on leukocytes during an immune response and suggest therapeutic interventions for optimizing immunity.
Collapse
Affiliation(s)
- Prajakta Shinde
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Alexander Kiepas
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lei Zhang
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shreya Sudhir
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Nicholas M Stamatos
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
8
|
Hunter C, Gao Z, Chen HM, Thompson N, Wakarchuk W, Nitz M, Withers SG, Willis LM. Attenuation of Polysialic Acid Biosynthesis in Cells by the Small Molecule Inhibitor 8-Keto-sialic acid. ACS Chem Biol 2023; 18:41-48. [PMID: 36577399 DOI: 10.1021/acschembio.2c00638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Sialic acids are key mediators of cell function, particularly with regard to cellular interactions with the surrounding environment. Reagents that modulate the display of specific sialyl glycoforms at the cell surface would be useful biochemical tools and potentially allow for therapeutic intervention in numerous challenging chronic diseases. While multiple strategies are being explored for the control of cell surface sialosides, none that shows high selectivity between sialyltransferases or that targets a specific sialyl glycoform has yet to emerge. Here, we describe a strategy to block the formation of α2,8-linked sialic acid chains (oligo- and polysialic acid) through the use of 8-keto-sialic acid as a chain-terminating metabolic inhibitor that, if incorporated, cannot be elongated. 8-Keto-sialic acid is nontoxic at effective concentrations and serves to block polysialic acid synthesis in cancer cell lines and primary immune cells, with minimal effects on other sialyl glycoforms.
Collapse
Affiliation(s)
- Carmanah Hunter
- Department of Biological Sciences, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Zhizeng Gao
- Department of Chemistry, University of British Columbia, Vancouver, V6T 1Z1, Canada
| | - Hong-Ming Chen
- Department of Chemistry, University of Toronto, Toronto, M5S 3H6, Canada
| | - Nicole Thompson
- Department of Biological Sciences, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Warren Wakarchuk
- Department of Biological Sciences, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Mark Nitz
- Department of Chemistry, University of Toronto, Toronto, M5S 3H6, Canada
| | - Stephen G Withers
- Department of Chemistry, University of British Columbia, Vancouver, V6T 1Z1, Canada
| | - Lisa M Willis
- Department of Biological Sciences, University of Alberta, Edmonton, T6G 2R3, Canada
| |
Collapse
|
9
|
Dhupar R, Jones KE, Powers AA, Eisenberg SH, Ding K, Chen F, Nasarre C, Cen Z, Gong YN, LaRue AC, Yeh ES, Luketich JD, Lee AV, Oesterreich S, Lotze MT, Gemmill RM, Soloff AC. Isoforms of Neuropilin-2 Denote Unique Tumor-Associated Macrophages in Breast Cancer. Front Immunol 2022; 13:830169. [PMID: 35651620 PMCID: PMC9149656 DOI: 10.3389/fimmu.2022.830169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor-associated macrophages (TAMs) exert profound influence over breast cancer progression, promoting immunosuppression, angiogenesis, and metastasis. Neuropilin-2 (NRP2), consisting of the NRP2a and NRP2b isoforms, is a co-receptor for heparin-binding growth factors including VEGF-C and Class 3 Semaphorins. Selective upregulation in response to environmental stimuli and independent signaling pathways endow the NRP2 isoforms with unique functionality, with NRP2b promoting increased Akt signaling via receptor tyrosine kinases including VEGFRs, MET, and PDGFR. Although NRP2 has been shown to regulate macrophage/TAM biology, the role of the individual NRP2a/NRP2b isoforms in TAMs has yet to be evaluated. Using transcriptional profiling and spectral flow cytometry, we show that NRP2 isoform expression was significantly higher in TAMs from murine mammary tumors. NRP2a/NRP2b levels in human breast cancer metastasis were dependent upon the anatomic location of the tumor and significantly correlated with TAM infiltration in both primary and metastatic breast cancers. We define distinct phenotypes of NRP2 isoform-expressing TAMs in mouse models of breast cancer and within malignant pleural effusions from breast cancer patients which were exclusive of neuropilin-1 expression. Genetic depletion of either NRP2 isoform in macrophages resulted in a dramatic reduction of LPS-induced IL-10 production, defects in phagosomal processing of apoptotic breast cancer cells, and increase in cancer cell migration following co-culture. By contrast, depletion of NRP2b, but not NRP2a, inhibited production of IL-6. These results suggest that NRP2 isoforms regulate both shared and unique functionality in macrophages and are associated with distinct TAM subsets in breast cancer.
Collapse
Affiliation(s)
- Rajeev Dhupar
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Surgical Services Division, VA Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - Katherine E Jones
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Amy A Powers
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Seth H Eisenberg
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Kai Ding
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee Women's Research Institute, Pittsburgh, PA, United States
| | - Fangyuan Chen
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee Women's Research Institute, Pittsburgh, PA, United States
| | - Cecile Nasarre
- Division of Hematology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
- Division of Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Zhanpeng Cen
- Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- School of Medicine, Tsinghua University, Beijing, China
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yi-Nan Gong
- Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Amanda C LaRue
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States
- Research Service, Ralph H. Johnson VA Health Care System, Charleston, SC, United States
| | - Elizabeth S Yeh
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Simon Cancer Center, Indianapolis, IN, United States
| | - James D Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Adrian V Lee
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee Women's Research Institute, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee Women's Research Institute, Pittsburgh, PA, United States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael T Lotze
- Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Robert M Gemmill
- Division of Hematology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
- Division of Oncology, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Adam C Soloff
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
- Research Service, Ralph H. Johnson VA Health Care System, Charleston, SC, United States
| |
Collapse
|
10
|
Immormino RM, Jania CM, Tilley SL, Moran TP. Neuropilin‐2 regulates airway inflammation in a neutrophilic asthma model. Immun Inflamm Dis 2022; 10:e575. [PMID: 34861108 PMCID: PMC8926497 DOI: 10.1002/iid3.575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/03/2021] [Accepted: 11/19/2021] [Indexed: 01/22/2023] Open
Abstract
Background Asthma is a heterogenous disease that can be classified into eosinophilic (type 2‐high) and noneosinophilic (type 2‐low) endotypes. The type 2‐low endotype of asthma can be characterized by the presence of neutrophilic airway inflammation that is poorly responsive to corticosteroids. Dysregulated innate immune responses to microbial products including Toll‐like receptor (TLR) ligands have been associated with the pathogenesis of neutrophilic asthma. The key molecules that regulate inflammatory responses in individuals with neutrophilic asthma remain unclear. We previously reported that the immunoregulatory receptor neuropilin‐2 (NRP2) is expressed by murine and human alveolar macrophage (AM) and suppresses lipopolysaccharide (LPS)‐induced neutrophilic airway inflammation. Methods Here, we investigated the immunoregulatory role of NRP2 in a mouse model of neutrophilic asthma. Results We found that TLR ligands, but not T helper 2 (Th2)‐promoting adjuvants, induced NRP2 expression by AM. Using an LPS‐mediated model of neutrophilic asthma, we demonstrate that NRP2 was increased in AM and other lung antigen‐presenting cells following airway challenge with antigen. Conditional deletion of NRP2 in myeloid cells exacerbated airway inflammation in a neutrophilic asthma model. In contrast, myeloid‐specific ablation of NRP2 did not affect airway inflammation in a Th2‐mediated eosinophilic asthma model. Myeloid‐specific ablation of NRP2 did not affect Th1/Th17 responses to inhaled antigens or expression of neutrophil chemokines but rather resulted in impaired efferocytosis by AM, which is necessary for effective resolution of airway inflammation. Conclusion Our findings suggest that NRP2 is a negative regulator of airway inflammation associated with neutrophilic asthma.
Collapse
Affiliation(s)
- Robert M. Immormino
- Center for Environmental Medicine, Asthma and Lung Biology University of North Carolina Chapel Hill North Carolina USA
| | - Corey M. Jania
- Department of Medicine University of North Carolina Chapel Hill North Carolina USA
| | - Stephen L. Tilley
- Department of Medicine University of North Carolina Chapel Hill North Carolina USA
| | - Timothy P. Moran
- Center for Environmental Medicine, Asthma and Lung Biology University of North Carolina Chapel Hill North Carolina USA
- Department of Pediatrics University of North Carolina Chapel Hill North Carolina USA
| |
Collapse
|
11
|
Villanueva-Cabello TM, Gutiérrez-Valenzuela LD, Salinas-Marín R, López-Guerrero DV, Martínez-Duncker I. Polysialic Acid in the Immune System. Front Immunol 2022; 12:823637. [PMID: 35222358 PMCID: PMC8873093 DOI: 10.3389/fimmu.2021.823637] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 12/28/2021] [Indexed: 01/01/2023] Open
Abstract
Polysialic acid (polySia) is a highly regulated polymer of sialic acid (Sia) with such potent biophysical characteristics that when expressed drastically influences the interaction properties of cells. Although much of what is known of polySia in mammals has been elucidated from the study of its role in the central nervous system (CNS), polySia is also expressed in other tissues, including the immune system where it presents dynamic changes during differentiation, maturation, and activation of different types of immune cells of the innate and adaptive response, being involved in key regulatory mechanisms. At least six polySia protein carriers (CCR7, ESL-1, NCAM, NRP2, ST8Sia 2, and ST8Sia 4) are expressed in different types of immune cells, but there is still much to be explored in regard not only to the regulatory mechanisms that determine their expression and the structure of polySia chains but also to the identification of the cis- and trans- ligands of polySia that establish signaling networks. This review summarizes the current knowledge on polySia in the immune system, addressing its biosynthesis, its tools for identification and structural characterization, and its functional roles and therapeutic implications.
Collapse
Affiliation(s)
- Tania M. Villanueva-Cabello
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Lya D. Gutiérrez-Valenzuela
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
- Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Roberta Salinas-Marín
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | | | - Iván Martínez-Duncker
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
- *Correspondence: Iván Martínez-Duncker,
| |
Collapse
|
12
|
Mindler K, Ostertag E, Stehle T. The polyfunctional polysialic acid: A structural view. Carbohydr Res 2021; 507:108376. [PMID: 34273862 DOI: 10.1016/j.carres.2021.108376] [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: 03/04/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 12/15/2022]
Abstract
Polysialic acid (polySia), a homopolymer of α2,8-linked sialic acid residues, modifies a small number of proteins and has central functions in vertebrate signalling. Here, we review the regulatory functions of polySia in signalling processes and the immune system of adult humans, as well as functions based on their chemical properties. The main focus will be on the structure-function relationship of polySia with its interaction partners in humans. Recent studies have indicated that the degree of polymerisation is an important parameter that can guide the regulatory effect of polySia in addition to its binding to target proteins. Therefore, the structures of polySia in solution and bound to interaction partners are compared in order to identify the key factors that define binding specificity.
Collapse
Affiliation(s)
- Katja Mindler
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076, Tübingen, Germany
| | - Elena Ostertag
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076, Tübingen, Germany
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076, Tübingen, Germany.
| |
Collapse
|
13
|
Harman JL, Sayers J, Chapman C, Pellet-Many C. Emerging Roles for Neuropilin-2 in Cardiovascular Disease. Int J Mol Sci 2020; 21:E5154. [PMID: 32708258 PMCID: PMC7404143 DOI: 10.3390/ijms21145154] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease, the leading cause of death worldwide, is predominantly associated with atherosclerosis. Atherosclerosis is a chronic inflammatory disease characterised by the narrowing of large to medium-sized arteries due to a build-up of plaque. Atherosclerotic plaque is comprised of lipids, extracellular matrix, and several cell types, including endothelial, immune, and vascular smooth muscle cells. Such narrowing of the blood vessels can itself restrict blood flow to vital organs but most severe clinical complications, including heart attacks and strokes, occur when lesions rupture, triggering the blood to clot and obstructing blood flow further down the vascular tree. To circumvent such obstructions, percutaneous coronary intervention or bypass grafts are often required; however, re-occlusion of the treated artery frequently occurs. Neuropilins (NRPs), a multifunctional family of cell surface co-receptors, are expressed by endothelial, immune, and vascular smooth muscle cells and are regulators of numerous signalling pathways within the vasculature. Here, we review recent studies implicating NRP2 in the development of occlusive vascular diseases and discuss how NRP2 could be targeted for therapeutic intervention.
Collapse
Affiliation(s)
- Jennifer L Harman
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Jacob Sayers
- University College London, Division of Medicine, Rayne Building, University Street, London WC1E 6JF, UK
| | - Chey Chapman
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| | - Caroline Pellet-Many
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK
| |
Collapse
|
14
|
Dumond A, Pagès G. Neuropilins, as Relevant Oncology Target: Their Role in the Tumoral Microenvironment. Front Cell Dev Biol 2020; 8:662. [PMID: 32766254 PMCID: PMC7380111 DOI: 10.3389/fcell.2020.00662] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/01/2020] [Indexed: 12/18/2022] Open
Abstract
Angiogenesis is one of the key mechanisms involved in tumor growth and metastatic dissemination. The vascular endothelial growth factor (VEGF) and its receptors (VEGFR) represent one of the major signaling pathways which mediates angiogenesis. The VEGF/VEGFR axis was intensively targeted by monoclonal antibodies or by tyrosine kinase inhibitors to destroy the tumor vascular network. By inhibiting oxygen and nutrient supply, this strategy was supposed to cure cancers. However, despite a lengthening of the progression free survival in several types of tumors including colon, lung, breast, kidney, and ovarian cancers, modest improvements in overall survival were reported. Anti-angiogenic therapies targeting VEGF/VEGFR are still used in colon and ovarian cancer and remain reference treatments for renal cell carcinoma. Although the concept of inhibiting angiogenesis remains relevant, new targets need to be discovered to improve the therapeutic index of anti-VEGF/VEGFR. Neuropilin 1 and 2 (NRP1/2), initially described as neuronal receptors, stimulate angiogenesis, lymphangiogenesis and immune tolerance. Moreover, overexpression of NRPs in several tumors is synonymous of patients' shorter survival. This article aims to overview the different roles of NRPs in cells constituting the tumor microenvironment to highlight the therapeutic relevance of their targeting.
Collapse
Affiliation(s)
- Aurore Dumond
- Medical Biology Department, Centre Scientifique de Monaco, Monaco, Monaco
| | - Gilles Pagès
- Medical Biology Department, Centre Scientifique de Monaco, Monaco, Monaco.,Inserm U1081, CNRS UMR 7284, Centre Antoine Lacassagne, Institut de Recherche sur le Cancer et le Vieillissement de Nice, Université Côte d'Azur, Nice, France
| |
Collapse
|
15
|
Tajik A, Phillips KL, Nitz M, Willis LM. A new ELISA assay demonstrates sex differences in the concentration of serum polysialic acid. Anal Biochem 2020; 600:113743. [DOI: 10.1016/j.ab.2020.113743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/01/2020] [Accepted: 04/14/2020] [Indexed: 12/20/2022]
|
16
|
Dumond A, Demange L, Pagès G. [Neuropilins: relevant therapeutic targets to improve the treatment of cancers]. Med Sci (Paris) 2020; 36:487-496. [PMID: 32452371 DOI: 10.1051/medsci/2020080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Exacerbated angiogenesis is one of the hallmarks of cancer defined by Hanahan and Weinberg. However, targeting the signaling pathway of the "Vascular Endothelial Growth Factor (VEGF)" or its receptors has shown its therapeutic limits. Despite short term benefits for patients, tumors always relapse and generally become metastatic and incurable. Neuropilins 1 and 2 (NRP1, 2) whose activity was originally described in the nervous system, stimulate many parameters involved in tumor aggressiveness including cell proliferation, angiogenesis and lymphangiogenesis, and immune tolerance. Thus, an overexpression of NRP1 or 2 in many tumors, is correlated with a short survival of the patients. The purpose of this review is to describe the mechanisms of action involved in stimulating NRP1, 2 and to take stock of therapeutic strategies in preclinical studies or in early phase trials in patients with different cancers.
Collapse
Affiliation(s)
- Aurore Dumond
- Centre scientifique de Monaco, Département de biologie médicale, 8 quai Antoine Ier, MC-98000 Monaco, Principauté de Monaco
| | - Luc Demange
- Université de Paris, CiTCoM, UMR 8038 CNRS, Faculté de Pharmacie, 4 avenue de l'Observatoire, F-75006 Paris, France
| | - Gilles Pagès
- Centre scientifique de Monaco, Département de biologie médicale, 8 quai Antoine Ier, MC-98000 Monaco, Principauté de Monaco - Université Côte d'Azur, Institut de recherche sur le cancer et le vieillissement de Nice, CNRS UMR 7284 ; Inserm U1081, Centre Antoine Lacassagne, 33 avenue de Valombrose, 06189 Nice, France
| |
Collapse
|
17
|
Wang W, Gopal S, Pocock R, Xiao Z. Glycan Mimetics from Natural Products: New Therapeutic Opportunities for Neurodegenerative Disease. Molecules 2019; 24:molecules24244604. [PMID: 31888221 PMCID: PMC6943557 DOI: 10.3390/molecules24244604] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/20/2022] Open
Abstract
Neurodegenerative diseases (NDs) affect millions of people worldwide. Characterized by the functional loss and death of neurons, NDs lead to symptoms (dementia and seizures) that affect the daily lives of patients. In spite of extensive research into NDs, the number of approved drugs for their treatment remains limited. There is therefore an urgent need to develop new approaches for the prevention and treatment of NDs. Glycans (carbohydrate chains) are ubiquitous, abundant, and structural complex natural biopolymers. Glycans often covalently attach to proteins and lipids to regulate cellular recognition, adhesion, and signaling. The importance of glycans in both the developing and mature nervous system is well characterized. Moreover, glycan dysregulation has been observed in NDs such as Alzheimer's disease (AD), Huntington's disease (HD), Parkinson's disease (PD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS). Therefore, glycans are promising but underexploited therapeutic targets. In this review, we summarize the current understanding of glycans in NDs. We also discuss a number of natural products that functionally mimic glycans to protect neurons, which therefore represent promising new therapeutic approaches for patients with NDs.
Collapse
|
18
|
Żaczek M, Górski A, Skaradzińska A, Łusiak-Szelachowska M, Weber-Dąbrowska B. Phage penetration of eukaryotic cells: practical implications. Future Virol 2019. [DOI: 10.2217/fvl-2019-0110] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The inability to infect eukaryotic cells has been considered as the most undeniable feature of all bacterial viruses. Such specificity, limited only for bacterial hosts, raises questions about the paths and challenges phages should overcome when circulating through the human body. Recently, it has been shown that phages are able to continually penetrate human organs and tissues. Latest reports revealed that phages can cross eukaryotic cell barriers both para- and transcellularly and even reach the nucleus. Further, phages are capable of internalizing within cells through different endocytic mechanisms. Such phenomenon indicates that phages could shape human microbiome composition and affect all aspects of human health. Thus, herein, we summarize the current state of knowledge and describe this phenomenon with a particular emphasis on endocytic pathways.
Collapse
Affiliation(s)
- Maciej Żaczek
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
| | - Andrzej Górski
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
- Phage Therapy Unit, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
| | - Aneta Skaradzińska
- Department of Biotechnology & Food Microbiology, Faculty of Biotechnology & Food Science, Wrocław University of Environmental & Life Sciences, Chełmońskiego 37, 51-630 Wrocław, Poland
| | - Marzanna Łusiak-Szelachowska
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
| | - Beata Weber-Dąbrowska
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
- Phage Therapy Unit, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), R. Weigla 12, 53-114 Wrocław, Poland
| |
Collapse
|
19
|
Cox EC, Thornlow DN, Jones MA, Fuller JL, Merritt JH, Paszek MJ, Alabi CA, DeLisa MP. Antibody-Mediated Endocytosis of Polysialic Acid Enables Intracellular Delivery and Cytotoxicity of a Glycan-Directed Antibody-Drug Conjugate. Cancer Res 2019; 79:1810-1821. [PMID: 30808675 PMCID: PMC6467748 DOI: 10.1158/0008-5472.can-18-3119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/01/2019] [Accepted: 02/21/2019] [Indexed: 12/23/2022]
Abstract
The specific targeting of differentially expressed glycans in malignant cells has emerged as an attractive anticancer strategy. One such target is the oncodevelopmental antigen polysialic acid (polySia), a polymer of α2,8-linked sialic acid residues that is largely absent during postnatal development but is re-expressed during progression of several malignant human tumors, including small-cell and non-small cell lung carcinomas, glioma, neuroblastoma, and pancreatic carcinoma. In these cancers, expression of polySia correlates with tumor progression and poor prognosis and appears to modulate cancer cell adhesion, invasiveness, and metastasis. To evaluate the potential of PolySia as a target for anticancer therapy, we developed a chimeric human polySia-specific mAb that retained low nanomolar (nmol/L) target affinity and exhibited exquisite selectivity for polySia structures. The engineered chimeric mAb recognized several polySia-positive tumor cell lines in vitro and induced rapid endocytosis of polySia antigens. To determine whether this internalization could be exploited for delivery of conjugated cytotoxic drugs, we generated an antibody-drug conjugate (ADC) by covalently linking the chimeric human mAb to the tubulin-binding maytansinoid DM1 using a bioorthogonal chemical reaction scheme. The resulting polySia-directed ADC demonstrated potent target-dependent cytotoxicity against polySia-positive tumor cells in vitro. Collectively, these results establish polySia as a valid cell-surface, cancer-specific target for glycan-directed ADC and contribute to a growing body of evidence that the tumor glycocalyx is a promising target for synthetic immunotherapies. SIGNIFICANCE: These findings describe a glycan-specific antibody-drug conjugate that establishes polySia as a viable cell surface target within the tumor glycocalyx.
Collapse
Affiliation(s)
- Emily C Cox
- Biological and Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York
| | - Dana N Thornlow
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| | - Michaela A Jones
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| | - Jordan L Fuller
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| | | | - Matthew J Paszek
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| | - Christopher A Alabi
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| | - Matthew P DeLisa
- Biological and Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York.
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York
| |
Collapse
|
20
|
Zhang X, Nie H, Whited J, Wang D, Li Y, Sun XL. Recent approaches for directly profiling cell surface sialoform. Glycobiology 2019; 28:910-924. [PMID: 29800278 DOI: 10.1093/glycob/cwy046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/09/2018] [Indexed: 12/13/2022] Open
Abstract
Sialic acids (SAs) are nine-carbon monosaccharides existing at the terminal location of glycan structures on the cell surface and secreted glycoconjugates. The expression levels and linkages of SAs on cells and tissues, collectively known as sialoform, present the hallmark of the cells and tissues of different systems and conditions. Accordingly, detecting or profiling cell surface sialoforms is very critical for understanding the function of cell surface glycans and glycoconjugates and even the molecular mechanisms of their underlying biological processes. Further, it may provide therapeutic and diagnostic applications for different diseases. In the past decades, several kinds of SA-specific binding molecules have been developed for detecting and profiling specific sialoforms of cells and tissues; the experimental materials have expanded from frozen tissue to living cells; and the analytical technologies have advanced from histochemistry to fluorescent imaging, flow cytometry and microarrays. This review summarizes the recent bioaffinity approaches for directly detecting and profiling specific SAs or sialylglycans, and their modifications of different cells and tissues.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- School of Life Science and Technology, Harbin Institute of Technology, 2 Yikuang-jie, Harbin, Heilongjiang, China
| | - Huan Nie
- School of Life Science and Technology, Harbin Institute of Technology, 2 Yikuang-jie, Harbin, Heilongjiang, China
| | - Joshua Whited
- Department of Chemistry, Chemical and Biomedical Engineering and Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, 2121 Euclid Avenue, Cleveland, OH, USA
| | - Dan Wang
- Department of Chemistry, Chemical and Biomedical Engineering and Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, 2121 Euclid Avenue, Cleveland, OH, USA
| | - Yu Li
- School of Life Science and Technology, Harbin Institute of Technology, 2 Yikuang-jie, Harbin, Heilongjiang, China
| | - Xue-Long Sun
- Department of Chemistry, Chemical and Biomedical Engineering and Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, 2121 Euclid Avenue, Cleveland, OH, USA
| |
Collapse
|
21
|
Paul A, Padler-Karavani V. Evolution of sialic acids: Implications in xenotransplant biology. Xenotransplantation 2018; 25:e12424. [PMID: 29932472 PMCID: PMC6756921 DOI: 10.1111/xen.12424] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/13/2022]
Abstract
All living cells are covered with a dense “sugar-coat” of carbohydrate chains (glycans) conjugated to proteins and lipids. The cell surface glycome is determined by a non-template driven process related to the collection of enzymes that assemble glycans in a sequential manner. In mammals, many of these glycans are topped with sialic acids (Sia), a large family of acidic sugars. The “Sialome” is highly diverse owing to various Sia types, linkage to underlying glycans, range of carriers, and complex spatial organization. Presented at the front of cells, Sia play a major role in immunity and recognition of “self” versus “non-self,” largely mediated by the siglecs family of Sia-binding host receptors. Albeit many mammalian pathogens have evolved to hijack this recognition system to avoid host immune attack, presenting a fascinating host-pathogen evolutionary arms race. Similarly, cancer cells exploit Sia for their own survival and propagation. As part of this ongoing fitness, humans lost the ability to synthesize the Sia type N-glycolylneuraminic acid (Neu5Gc), in contrast to other mammals. While this loss had provided an advantage against certain pathogens, humans are continuously exposed to Neu5Gc through mammalian-derived diet (eg, red meat), consequently generating a complex immune response against it. Circulating anti-Neu5Gc antibodies together with Neu5Gc on some human tissues mediate chronic inflammation “xenosialitis” that exacerbate various human diseases (eg, cancer and atherosclerosis). Similarly, Neu5Gc-containing xenografts are exposed to human anti-Neu5Gc antibodies with implications to sustainability. This review aimed to provide a glimpse into the evolution of Sia and their implications to xenotransplantation.
Collapse
Affiliation(s)
- Anu Paul
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Vered Padler-Karavani
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
22
|
Abstract
We present here that adenovirus type 52 (HAdV-52) attaches to target cells through a mechanism not previously observed in other human pathogenic viruses. The interaction involves unusual, transient, electrostatic interactions between the short fiber capsid protein and polysialic acid (polySia)-containing receptors on target cells. Knowledge about the binding interactions between polySia and its natural ligands is relatively limited, and our results therefore provide additional insight not only into adenovirus biology but also into the structural basis of polySia function. Since polySia can be found in high expression levels in brain and lung cancers where its presence is associated with poor prognosis, we suggest that this polySia-binding adenovirus could be useful for design of vectors for gene therapy of these cancers. Human adenovirus 52 (HAdV-52) is one of only three known HAdVs equipped with both a long and a short fiber protein. While the long fiber binds to the coxsackie and adenovirus receptor, the function of the short fiber in the virus life cycle is poorly understood. Here, we show, by glycan microarray analysis and cellular studies, that the short fiber knob (SFK) of HAdV-52 recognizes long chains of α-2,8-linked polysialic acid (polySia), a large posttranslational modification of selected carrier proteins, and that HAdV-52 can use polySia as a receptor on target cells. X-ray crystallography, NMR, molecular dynamics simulation, and structure-guided mutagenesis of the SFK reveal that the nonreducing, terminal sialic acid of polySia engages the protein with direct contacts, and that specificity for polySia is achieved through subtle, transient electrostatic interactions with additional sialic acid residues. In this study, we present a previously unrecognized role for polySia as a cellular receptor for a human viral pathogen. Our detailed analysis of the determinants of specificity for this interaction has general implications for protein–carbohydrate interactions, particularly concerning highly charged glycan structures, and provides interesting dimensions on the biology and evolution of members of Human mastadenovirus G.
Collapse
|
23
|
Individual Impact of Distinct Polysialic Acid Chain Lengths on the Cytotoxicity of Histone H1, H2A, H2B, H3 and H4. Polymers (Basel) 2017; 9:polym9120720. [PMID: 30966022 PMCID: PMC6418544 DOI: 10.3390/polym9120720] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 12/28/2022] Open
Abstract
Neutrophils are able to neutralize pathogens by phagocytosis, by the release of antimicrobial components, as well as by the formation of neutrophil extracellular traps (NETs). The latter possibility is a DNA-meshwork mainly consisting of highly concentrated extracellular histones, which are not only toxic for pathogens, but also for endogenous cells triggering several diseases. To reduce the negative outcomes initiated by extracellular histones, different approaches like antibodies against histones, proteases, and the polysaccharide polysialic acid (polySia) were discussed. We examined whether each of the individual histones is a binding partner of polySia, and analyzed their respective cytotoxicity in the presence of this linear homopolymer. Interestingly, all of the histones (H1, H2A, H2B, H3, and H4) seem to interact with α2,8-linked sialic acids. However, we observed strong differences regarding the required chain length of polySia to bind histone H1, H2A, H2B, H3, and H4. Moreover, distinct degrees of polymerization were necessary to act as a cytoprotective agent in the presence of the individual histones. In sum, the outlined results described polySia-based strategies to bind and/or to reduce the cytotoxicity of individual histones using distinct polySia chain length settings.
Collapse
|
24
|
Roy S, Bag AK, Singh RK, Talmadge JE, Batra SK, Datta K. Multifaceted Role of Neuropilins in the Immune System: Potential Targets for Immunotherapy. Front Immunol 2017; 8:1228. [PMID: 29067024 PMCID: PMC5641316 DOI: 10.3389/fimmu.2017.01228] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/19/2017] [Indexed: 12/27/2022] Open
Abstract
Neuropilins (NRPs) are non-tyrosine kinase cell surface glycoproteins expressed in all vertebrates and widely conserved across species. The two isoforms, such as neuropilin-1 (NRP1) and neuropilin-2 (NRP2), mainly act as coreceptors for class III Semaphorins and for members of the vascular endothelial growth factor family of molecules and are widely known for their role in a wide array of physiological processes, such as cardiovascular, neuronal development and patterning, angiogenesis, lymphangiogenesis, as well as various clinical disorders. Intriguingly, additional roles for NRPs occur with myeloid and lymphoid cells, in normal physiological as well as different pathological conditions, including cancer, immunological disorders, and bone diseases. However, little is known concerning the molecular pathways that govern these functions. In addition, NRP1 expression has been characterized in different immune cellular phenotypes including macrophages, dendritic cells, and T cell subsets, especially regulatory T cell populations. By contrast, the functions of NRP2 in immune cells are less well known. In this review, we briefly summarize the genomic organization, structure, and binding partners of the NRPs and extensively discuss the recent advances in their role and function in different immune cell subsets and their clinical implications.
Collapse
Affiliation(s)
- Sohini Roy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Arup K Bag
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Rakesh K Singh
- Department of Microbiology and Pathology, University of Nebraska Medical Center, Omaha, NE, United States
| | - James E Talmadge
- Department of Microbiology and Pathology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
25
|
Schellenburg S, Schulz A, Poitz DM, Muders MH. Role of neuropilin-2 in the immune system. Mol Immunol 2017; 90:239-244. [PMID: 28843905 DOI: 10.1016/j.molimm.2017.08.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/14/2017] [Indexed: 01/07/2023]
Abstract
Neuropilins (NRPs) are single transmembrane receptors with short cytoplasmic tails and are dependent on receptors like VEGF receptors or Plexins for signal transduction. NRPs are known to be important in angiogenesis, lymphangiogenesis, and axon guidance. The Neuropilin-family consists of two members, Neuropilin-1 (NRP1) and Neuropilin-2 (NRP2). They are up to 44 % homologous and conserved in all vertebrates. High levels of NRP2 are found on immune cells. Current research is very limited regarding the functions of NRP2 on these cells. Recent evidence suggests that NRP2 is important for migration, antigen presentation, phagocytosis and cell-cell contact within the immune system. Additionally, posttranslational NRP2 modifications like polysialylation are crucial for the function of some immune cells. This review is an overview about expression and functions of NRP2 in the immune system.
Collapse
Affiliation(s)
- S Schellenburg
- Institute of Pathology, University Hospital Carl Gustav Carus, University of Technology, Dresden, Germany
| | - A Schulz
- Institute of Pathology, University Hospital Carl Gustav Carus, University of Technology, Dresden, Germany
| | - D M Poitz
- Department of Internal Medicine and Cardiology, University Hospital Carl Gustav Carus, University of Technology, Dresden, Germany
| | - M H Muders
- Institute of Pathology, University Hospital Carl Gustav Carus, University of Technology, Dresden, Germany.
| |
Collapse
|
26
|
Galuska CE, Lütteke T, Galuska SP. Is Polysialylated NCAM Not Only a Regulator during Brain Development But also during the Formation of Other Organs? BIOLOGY 2017; 6:biology6020027. [PMID: 28448440 PMCID: PMC5485474 DOI: 10.3390/biology6020027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 11/16/2022]
Abstract
In mammals several cell adhesion molecules are involved during the pre- and postnatal development of all organ systems. A very prominent member of this family is the neural cell adhesion molecule (NCAM). Interestingly, NCAM can be a target for a special form of posttranslational modification: polysialylation. Whereas nearly all extracellular proteins bear mono-sialic acid residues, only a very small group can be polysialylated. Polysialic acid is a highly negatively-charged sugar polymer and can comprise more than 90 sialic acid residues in postnatal mouse brains increasing dramatically the hydrodynamic radius of their carriers. Thus, adhesion and communication processes on cell surfaces are strongly influenced allowing, e.g., the migration of neuronal progenitor cells. In the developing brain the essential role of polysialylated NCAM has been demonstrated in many studies. In comparison to the neuronal system, however, during the formation of other organs the impact of the polysialylated form of NCAM is not well characterized and the number of studies is limited so far. This review summarizes these observations and discusses possible roles of polysialylated NCAM during the development of organs other than the brain.
Collapse
Affiliation(s)
- Christina E Galuska
- Department of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| | - Thomas Lütteke
- ITech Progress GmbH, Donnersbergweg 4, 67059 Ludwigshafen, Germany.
| | - Sebastian P Galuska
- Department of Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| |
Collapse
|
27
|
Mehrabian M, Hildebrandt H, Schmitt-Ulms G. NCAM1 Polysialylation: The Prion Protein's Elusive Reason for Being? ASN Neuro 2016; 8:8/6/1759091416679074. [PMID: 27879349 PMCID: PMC5122176 DOI: 10.1177/1759091416679074] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 09/08/2016] [Accepted: 10/02/2016] [Indexed: 01/06/2023] Open
Abstract
Much confusion surrounds the physiological function of the cellular prion protein (PrPC). It is, however, anticipated that knowledge of its function will shed light on its contribution to neurodegenerative diseases and suggest ways to interfere with the cellular toxicity central to them. Consequently, efforts to elucidate its function have been all but exhaustive. Building on earlier work that uncovered the evolutionary descent of the prion founder gene from an ancestral ZIP zinc transporter, we recently investigated a possible role of PrPC in a morphogenetic program referred to as epithelial-to-mesenchymal transition (EMT). By capitalizing on PrPC knockout cell clones in a mammalian cell model of EMT and using a comparative proteomics discovery strategy, neural cell adhesion molecule-1 emerged as a protein whose upregulation during EMT was perturbed in PrPC knockout cells. Follow-up work led us to observe that PrPC regulates the polysialylation of the neural cell adhesion molecule NCAM1 in cells undergoing morphogenetic reprogramming. In addition to governing cellular migration, polysialylation modulates several other cellular plasticity programs PrPC has been phenotypically linked to. These include neurogenesis in the subventricular zone, controlled mossy fiber sprouting and trimming in the hippocampal formation, hematopoietic stem cell renewal, myelin repair and maintenance, integrity of the circadian rhythm, and glutamatergic signaling. This review revisits this body of literature and attempts to present it in light of this novel contextual framework. When approached in this manner, a coherent model of PrPC acting as a regulator of polysialylation during specific cell and tissue morphogenesis events comes into focus.
Collapse
Affiliation(s)
- Mohadeseh Mehrabian
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Herbert Hildebrandt
- Institute for Cellular Chemistry, Hannover Medical School, Hannover, Germany
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Curreli S, Wong BS, Latinovic O, Konstantopoulos K, Stamatos NM. Class 3 semaphorins induce F-actin reorganization in human dendritic cells: Role in cell migration. J Leukoc Biol 2016; 100:1323-1334. [PMID: 27406993 DOI: 10.1189/jlb.2a1114-534r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 05/12/2016] [Accepted: 06/08/2016] [Indexed: 01/20/2023] Open
Abstract
Class 3 semaphorins (Semas) are soluble proteins that are well recognized for their role in guiding axonal migration during neuronal development. In the immune system, Sema3A has been shown to influence murine dendritic cell (DC) migration by signaling through a neuropilin (NRP)-1/plexin-A1 coreceptor axis. Potential roles for class 3 Semas in human DCs have yet to be described. We tested the hypothesis that Sema3A, -3C, and -3F, each with a unique NRP-1 and/or NRP-2 binding specificity, influence human DC migration. In this report, we find that although NRP-1 and NRP-2 are expressed in human immature DCs (imDCs), NRP-2 expression increases as cells mature further, whereas expression of NRP-1 declines dramatically. Elevated levels of RNA encoding plexin-A1 and -A3 are present in both imDCs and mature DC (mDCs), supporting the relevance of Sema/NRP/plexin signaling pathways in these cells. Sema3A, -3C, and -3F bind to human DCs, with Sema3F binding predominantly through NRP-2. The binding of these Semas leads to reorganization of actin filaments at the plasma membrane and increased transwell migration in the absence or presence of chemokine CCL19. Microfluidic chamber assays failed to demonstrate consistent changes in speed of Sema3C-treated DCs, suggesting increased cell deformability as a possible explanation for enhanced transwell migration. Although monocytes express RNA encoding Sema3A, -3C, and -3F, only RNA encoding Sema3C increases robustly during DC differentiation. These data suggest that Sema3A, -3C, and -3F, likely with coreceptors NRP-1, NRP-2, and plexin-A1 and/or -A3, promote migration and possibly other activities of human DCs during innate and adaptive immune responses.
Collapse
Affiliation(s)
- Sabrina Curreli
- Institute of Human Virology, University of Maryland Medical Center, Baltimore, Maryland, USA
| | - Bin Sheng Wong
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Olga Latinovic
- Institute of Human Virology, University of Maryland Medical Center, Baltimore, Maryland, USA.,Department of Microbiology and Immunology, University of Maryland Medical Center, Baltimore, Maryland, USA
| | | | - Nicholas M Stamatos
- Institute of Human Virology, University of Maryland Medical Center, Baltimore, Maryland, USA; .,Department of Medicine, University of Maryland Medical Center, Baltimore, Maryland, USA; and
| |
Collapse
|
29
|
Werneburg S, Buettner FFR, Erben L, Mathews M, Neumann H, Mühlenhoff M, Hildebrandt H. Polysialylation and lipopolysaccharide-induced shedding of E-selectin ligand-1 and neuropilin-2 by microglia and THP-1 macrophages. Glia 2016; 64:1314-30. [DOI: 10.1002/glia.23004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/19/2016] [Accepted: 04/22/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Sebastian Werneburg
- Institute for Cellular Chemistry, Hannover Medical School; Carl-Neuberg-Straße 1 30625 Hannover Germany
- Center for Systems Neuroscience (ZSN); Hannover, Bünteweg 2 30559 Hannover Germany
| | - Falk F. R. Buettner
- Institute for Cellular Chemistry, Hannover Medical School; Carl-Neuberg-Straße 1 30625 Hannover Germany
| | - Larissa Erben
- Institute for Cellular Chemistry, Hannover Medical School; Carl-Neuberg-Straße 1 30625 Hannover Germany
| | - Mona Mathews
- Institute of Reconstructive Neurobiology, University of Bonn; Sigmund-Freud-Straße 25 53127 Bonn Germany
| | - Harald Neumann
- Institute of Reconstructive Neurobiology, University of Bonn; Sigmund-Freud-Straße 25 53127 Bonn Germany
| | - Martina Mühlenhoff
- Institute for Cellular Chemistry, Hannover Medical School; Carl-Neuberg-Straße 1 30625 Hannover Germany
| | - Herbert Hildebrandt
- Institute for Cellular Chemistry, Hannover Medical School; Carl-Neuberg-Straße 1 30625 Hannover Germany
- Center for Systems Neuroscience (ZSN); Hannover, Bünteweg 2 30559 Hannover Germany
| |
Collapse
|
30
|
Bhide GP, Fernandes NRJ, Colley KJ. Sequence Requirements for Neuropilin-2 Recognition by ST8SiaIV and Polysialylation of Its O-Glycans. J Biol Chem 2016; 291:9444-57. [PMID: 26884342 DOI: 10.1074/jbc.m116.714329] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Indexed: 01/05/2023] Open
Abstract
Polysialic acid is an oncofetal glycopolymer, added to the glycans of a small group of substrates, that controls cell adhesion and signaling. One of these substrates, neuropilin-2, is a VEGF and semaphorin co-receptor that is polysialylated on its O-glycans in mature dendritic cells and macrophages by the polysialyltransferase ST8SiaIV. To understand the biochemical basis of neuropilin-2 polysialylation, we created a series of domain swap chimeras with sequences from neuropilin-1, a protein for which polysialylation had not been previously reported. To our surprise, we found that membrane-associated neuropilin-1 is polysialylated at ∼50% of the level of neuropilin-2 but not polysialylated when it lacks its cytoplasmic tail and transmembrane region and is secreted from the cell. This was not the case for neuropilin-2, which is polysialylated when either membrane-associated or soluble. Evaluation of the soluble chimeric proteins demonstrated that the meprin A5 antigen-μ tyrosine phosphatase (MAM) domain and the O-glycan-containing linker region of neuropilin-2 are necessary and sufficient for its polysialylation and serve as better recognition and acceptor sites in the polysialylation process than those regions of neuropilin-1. In addition, specific acidic residues on the surface of the MAM domain are critical for neuropilin-2 polysialylation. Based on these data and pull-down experiments, we propose a model where ST8SiaIV recognizes and docks on an acidic surface of the neuropilin-2 MAM domain to polysialylate O-glycans on the adjacent linker region. These results together with those related to neural cell adhesion molecule polysialylation establish a paradigm for the process of protein-specific polysialylation.
Collapse
Affiliation(s)
- Gaurang P Bhide
- From the Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois 60607
| | - Ninoshka R J Fernandes
- From the Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois 60607
| | - Karen J Colley
- From the Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois 60607
| |
Collapse
|
31
|
Polysialic acid biosynthesis and production in Escherichia coli: current state and perspectives. Appl Microbiol Biotechnol 2015; 100:1-8. [DOI: 10.1007/s00253-015-7019-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 09/15/2015] [Accepted: 09/20/2015] [Indexed: 11/26/2022]
|
32
|
Kaese M, Galuska CE, Simon P, Braun BC, Cabrera-Fuentes HA, Middendorff R, Wehrend A, Jewgenow K, Galuska SP. Polysialylation takes place in granulosa cells during apoptotic processes of atretic tertiary follicles. FEBS J 2015; 282:4595-606. [PMID: 26392163 DOI: 10.1111/febs.13519] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 08/14/2015] [Accepted: 09/16/2015] [Indexed: 11/30/2022]
Abstract
In the neuronal system, polysialic acid (polySia) is known to be involved in several cellular processes such as the modulation of cell-cell interactions. This highly negatively-charged sugar moiety is mainly present as a post-translational modification of the neural cell adhesion molecule (NCAM). More than 20 years ago, differently glycosylated forms of NCAM were detected in the ovaries. However, the exact isoform of NCAM, as well as its biological function, remained unknown. Our analysis revealed that granulosa cells of feline tertiary follicles express the polysialylated form of NCAM-140. Unexpectedly, polySia was only expressed in the granulosa layers of atretic follicles and not of healthy follicles. By contrast, only the un-polysialylated form of NCAM was present on the membrane of granulosa cells of healthy follicles. To study a possible cellular function of polySia in feline follicles, a primary granulosa cell culture model was used. Interestingly, loss of polySia leads to a significant inhibition of apoptosis, demonstrating that polySia is involved during atretic processes in granulosa cells. Thus, polySia might not only directly influence regeneration processes as shown, for example, in the neuronal system, but also apoptosis.
Collapse
Affiliation(s)
- Miriam Kaese
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Giessen, Germany
| | - Christina E Galuska
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Giessen, Germany
| | - Peter Simon
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Giessen, Germany.,Institute of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Giessen, Germany
| | - Beate C Braun
- Leibniz Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
| | - Hector A Cabrera-Fuentes
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Giessen, Germany.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore
| | - Ralf Middendorff
- Institute of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Giessen, Germany
| | - Axel Wehrend
- Clinic of Obstetrics, Gynecology and Andrology for Small and Large Animals, Justus-Liebig-University, Giessen, Germany
| | - Katarina Jewgenow
- Leibniz Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
| | - Sebastian P Galuska
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
33
|
Simon P, Feuerstacke C, Kaese M, Saboor F, Middendorff R, Galuska SP. Polysialylation of NCAM characterizes the proliferation period of contractile elements during postnatal development of the epididymis. PLoS One 2015; 10:e0123960. [PMID: 25822229 PMCID: PMC4379024 DOI: 10.1371/journal.pone.0123960] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/11/2015] [Indexed: 11/19/2022] Open
Abstract
Polysialic acid (polySia) attached to the neural cell adhesion molecule (NCAM) regulates inter alia the proliferation and differentiation via the interactions with neurotrophins. Since in postnatal epididymis neurotrophins and their receptors like the Low-Affinity Nerve Growth Factor Receptor p75 and TrK B receptor are expressed, we wanted to analyze if the polysialylation of NCAM is also involved during the development of the epididymis. To this end, we monitored the developmental changes in the expression of the polysialyltransferases and NCAM polysialylation using murine epididymis at different time points during postnatal development. Our results revealed that during postnatal development of the epididymis both polysialyltransferases, ST8SiaII and ST8SiaIV, were expressed and that the expression levels dropped with increasing age. In agreement with the expression levels of the polysialyltransferases the highest content of polysialylated NCAM was present during the first 10 days after birth. Interestingly, proliferating smooth muscle cell populations prevalently expressed polysialylated NCAM. Furthermore, we observed that inverse to the decrease in polysialylation of smooth muscle cells a strong up-regulation of collagen takes place suggesting a functional relationship since collagen was recently described to induce the turnover of polysialylated NCAM via an induction of endocytosis in cellulo. The same time course of polySia and collagen synthesis was also observed in other regions of the male reproductive system e.g. vas deferens and tunica albuginea (testis). Together, we identified a spatio-temporal expression pattern of polySia-NCAM characterized by high proliferation rate of smooth muscle cells and low collagen content.
Collapse
Affiliation(s)
- Peter Simon
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Friedrichstr. 24, 35392, Giessen, Germany
- Department of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Aulweg 123, 35385, Giessen, Germany
| | - Caroline Feuerstacke
- Department of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Aulweg 123, 35385, Giessen, Germany
| | - Miriam Kaese
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Friedrichstr. 24, 35392, Giessen, Germany
| | - Farhan Saboor
- Department of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Aulweg 123, 35385, Giessen, Germany
| | - Ralf Middendorff
- Department of Anatomy and Cell Biology, Medical Faculty, Justus-Liebig-University, Aulweg 123, 35385, Giessen, Germany
- * E-mail: (RM); (SPG)
| | - Sebastian P. Galuska
- Institute of Biochemistry, Medical Faculty, Justus-Liebig-University, Friedrichstr. 24, 35392, Giessen, Germany
- * E-mail: (RM); (SPG)
| |
Collapse
|
34
|
Werneburg S, Mühlenhoff M, Stangel M, Hildebrandt H. Polysialic acid on SynCAM 1 in NG2 cells and on neuropilin-2 in microglia is confined to intracellular pools that are rapidly depleted upon stimulation. Glia 2015; 63:1240-55. [DOI: 10.1002/glia.22815] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/20/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Sebastian Werneburg
- Hannover Medical School; Institute for Cellular Chemistry; Carl-Neuberg-Straße 1 Hannover Germany
- Center for Systems Neuroscience (ZSN); Hannover Germany
| | - Martina Mühlenhoff
- Hannover Medical School; Institute for Cellular Chemistry; Carl-Neuberg-Straße 1 Hannover Germany
| | - Martin Stangel
- Center for Systems Neuroscience (ZSN); Hannover Germany
- Clinical Neuroimmunology and Neurochemistry; Department of Neurology; Hannover Medical School; Carl-Neuberg-Straße 1 Hannover Germany
| | - Herbert Hildebrandt
- Hannover Medical School; Institute for Cellular Chemistry; Carl-Neuberg-Straße 1 Hannover Germany
- Center for Systems Neuroscience (ZSN); Hannover Germany
| |
Collapse
|
35
|
Abstract
Oligo- and polysialic acids (oligo/polySia) are involved in numerous biological processes depending on the chain length, the comprised type of sialic acids, as well as the glycosidic linkages. Here, we describe the determination of the composition, the sequence, as well as the linkages between the sialic acid residues of lactonized oligo/polySia using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS)(/MS) and electrospray-ionization (ESI)-MS((n)).
Collapse
Affiliation(s)
- Christina E Galuska
- Institute of Biochemistry, Faculty of Medicine, University of Giessen, Friedrichstrasse 24, 35392, Giessen, Germany
| | | | | |
Collapse
|
36
|
Colley KJ, Kitajima K, Sato C. Polysialic acid: biosynthesis, novel functions and applications. Crit Rev Biochem Mol Biol 2014; 49:498-532. [PMID: 25373518 DOI: 10.3109/10409238.2014.976606] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
As an anti-adhesive, a reservoir for key biological molecules, and a modulator of signaling, polysialic acid (polySia) is critical for nervous system development and maintenance, promotes cancer metastasis, tissue regeneration and repair, and is implicated in psychiatric diseases. In this review, we focus on the biosynthesis and functions of mammalian polySia, and the use of polySia in therapeutic applications. PolySia modifies a small subset of mammalian glycoproteins, with the neural cell adhesion molecule, NCAM, serving as its major carrier. Studies show that mammalian polysialyltransferases employ a unique recognition mechanism to limit the addition of polySia to a select group of proteins. PolySia has long been considered an anti-adhesive molecule, and its impact on cell adhesion and signaling attributed directly to this property. However, recent studies have shown that polySia specifically binds neurotrophins, growth factors, and neurotransmitters and that this binding depends on chain length. This work highlights the importance of considering polySia quality and quantity, and not simply its presence or absence, as its various roles are explored. The capsular polySia of neuroinvasive bacteria allows these organisms to evade the host immune response. While this "stealth" characteristic has made meningitis vaccine development difficult, it has also made polySia a worthy replacement for polyetheylene glycol in the generation of therapeutic proteins with low immunogenicity and improved circulating half-lives. Bacterial polysialyltransferases are more promiscuous than the protein-specific mammalian enzymes, and new studies suggest that these enzymes have tremendous therapeutic potential, especially for strategies aimed at neural regeneration and tissue repair.
Collapse
Affiliation(s)
- Karen J Colley
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago , Chicago, IL , USA and
| | | | | |
Collapse
|