1
|
Schattner M, Psaila B, Rabinovich GA. Shaping hematopoietic cell ecosystems through galectin-glycan interactions. Semin Immunol 2024; 74-75:101889. [PMID: 39405834 DOI: 10.1016/j.smim.2024.101889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 11/18/2024]
Abstract
Hematopoiesis- the formation of blood cell components- continually replenishes the blood system during embryonic development and postnatal lifespans. This coordinated process requires the synchronized action of a broad range of cell surface associated proteins and soluble mediators, including growth factors, cytokines and lectins. Collectively, these mediators control cellular communication, signalling, commitment, proliferation, survival and differentiation. Here we discuss the role of galectins - an evolutionarily conserved family of glycan-binding proteins - in the establishment and dynamic remodelling of hematopoietic niches. We focus on the contribution of galectins to B and T lymphocyte development and selection, as well as studies highlighting the role of these proteins in myelopoiesis, with particular emphasis on erythropoiesis and megakaryopoiesis. Finally, we also highlight recent findings suggesting the role of galectin-1, a prototype member of this protein family, as a key pathogenic factor and therapeutic target in myelofibrosis. Through extracellular or intracellular mechanisms, galectins can influence the fate and function of distinct hematopoietic progenitors and fine-tune the final repertoire of blood cells, with critical implications in a wide range of physiologically vital processes including innate and adaptive immunity, immune tolerance programs, tissue repair, regeneration, angiogenesis, inflammation, coagulation and oxygen delivery. Additionally, positive or negative regulation of galectin-driven circuits may contribute to a broad range of blood cell disorders.
Collapse
Affiliation(s)
- Mirta Schattner
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad de Buenos Aires 1428, Argentina; Laboratorio de Trombosis Experimental e Inmunobiología de la Inflamación, Instituto de Medicina Experimental, CONICET-Academia Nacional de Medicina, Ciudad de Buenos Aires 1425, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires 1428, Argentina.
| | - Bethan Psaila
- MRC Weatherall Institute of Molecular Medicine and Ludwig Institute for Cancer Research, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad de Buenos Aires 1428, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires 1428, Argentina.
| |
Collapse
|
2
|
Querol Cano L, Dunlock VME, Schwerdtfeger F, van Spriel AB. Membrane organization by tetraspanins and galectins shapes lymphocyte function. Nat Rev Immunol 2024; 24:193-212. [PMID: 37758850 DOI: 10.1038/s41577-023-00935-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2023] [Indexed: 09/29/2023]
Abstract
Immune receptors are not randomly distributed at the plasma membrane of lymphocytes but are segregated into specialized domains that function as platforms to initiate signalling, as exemplified by the B cell or T cell receptor complex and the immunological synapse. 'Membrane-organizing proteins' and, in particular, tetraspanins and galectins, are crucial for controlling the spatiotemporal organization of immune receptors and other signalling proteins. Deficiencies in specific tetraspanins and galectins result in impaired immune synapse formation, lymphocyte proliferation, antibody production and migration, which can lead to impaired immunity, tumour development and autoimmunity. In contrast to conventional ligand-receptor interactions, membrane organizers interact in cis (on the same cell) and modulate receptor clustering, receptor dynamics and intracellular signalling. New findings have uncovered their complex and dynamic nature, revealing shared binding partners and collaborative activity in determining the composition of membrane domains. Therefore, immune receptors should not be envisaged as independent entities and instead should be studied in the context of their spatial organization in the lymphocyte membrane. We advocate for a novel approach to study lymphocyte function by globally analysing the role of membrane organizers in the assembly of different membrane complexes and discuss opportunities to develop therapeutic approaches that act via the modulation of membrane organization.
Collapse
Affiliation(s)
- Laia Querol Cano
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vera-Marie E Dunlock
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fabian Schwerdtfeger
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annemiek B van Spriel
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
3
|
Fryk E, Wilsson Å, Tompa A, Jansson PA, Faresjö M. Galectin-1 correlates with inflammatory markers and T regulatory cells in children with type 1 diabetes and/or celiac disease. Clin Exp Immunol 2024; 215:240-250. [PMID: 38088456 PMCID: PMC10876110 DOI: 10.1093/cei/uxad131] [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: 06/09/2023] [Revised: 11/01/2023] [Accepted: 12/05/2023] [Indexed: 02/20/2024] Open
Abstract
Type 1 diabetes (T1D) and celiac disease (CeD) are common autoimmune diseases in children where the pathophysiology is not fully characterized. The autoimmune process involves a complex scenario of both inflammatory and regulatory features. Galectin-1 (GAL-1) has a wide range of biological activities e.g. interaction with immune cells. We examined the relationship between GAL-1 and soluble immune markers and T-cell subsets in a cohort of children with T1D and/or CeD relative to healthy children. GAL-1, together with several soluble immune markers [e.g. interleukins (IL)], tumor necrosis factor (TNF), acute phase proteins, and matrix metalloproteinases (MMP) were measured in sera from children with T1D and/or CeD by fluorochrome (Luminex) technique using children without these diseases as a reference. Subgroups of T cells, including T-regulatory (Treg) cells, were analysed by flow cytometry. Association between GAL-1, pro-inflammatory markers, and Treg cells differed depending on which illness combination was present. In children with both T1D and CeD, GAL-1 correlated positively with pro-inflammatory markers (IL-1β, IL-6, and TNF-α). Composite scores increased the strength of correlation between GAL-1 and pro-inflammatory markers, Th1-associated interferon (IFN)-γ, and T1D-associated visfatin. Contrary, in children diagnosed with exclusively T1D, GAL-1 was positively correlated to CD25hi and CD25hiCD101+ Treg cells. For children with only CeD, no association between GAL-1 and other immune markers was observed. In conclusion, the association observed between GAL-1, soluble immune markers, and Treg cells may indicate a role for GAL-1 in the pathophysiology of T1D and, to some extent, also in CeD.
Collapse
Affiliation(s)
- Emanuel Fryk
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Åsa Wilsson
- Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Andrea Tompa
- Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
- Division of Medical Diagnostics, Department of Laboratory Medicine, Region Jönköping County, Sweden
| | - Per-Anders Jansson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Maria Faresjö
- Department of Life Sciences, Division of Systems and Synthetic Biology, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
4
|
Dam TK, Brewer CF. Multivalent lectin-carbohydrate interactions: Energetics and mechanisms of binding. Adv Carbohydr Chem Biochem 2023; 84:23-48. [PMID: 37979978 DOI: 10.1016/bs.accb.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2023]
Abstract
The biological signaling properties of lectins, which are carbohydrate-binding proteins, are due to their ability to bind and cross-link multivalent glycoprotein receptors on the surface of normal and transformed cells. While the cross-linking properties of lectins with multivalent carbohydrates and glycoproteins are relatively well understood, the mechanisms of binding of lectins to multivalent glycoconjugates are less well understood. Recently, the thermodynamics of binding of lectins to synthetic clustered glycosides, a multivalent globular glycoprotein, and to linear glycoproteins (mucins) have been described. The results are consistent with a dynamic binding mechanism in which lectins bind and jump from carbohydrate to carbohydrate epitope in these molecules. Importantly, the mechanism of binding of lectins to mucins is similar to that for a variety of protein ligands binding to DNA. Recent analysis also shows that high-affinity lectin-mucin cross-linking interactions are driven by favorable entropy of binding that is associated with the bind and jump mechanism. The results suggest that the binding of ligands to biopolymers, in general, may involve a common mechanism that involves enhanced entropic effects which facilitate binding and subsequent complex formation including enzymology.
Collapse
Affiliation(s)
- Tarun K Dam
- Formerly of the Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States.
| | - C Fred Brewer
- Department of Molecular Pharmacology, Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
5
|
Abdelbary M, Nolz JC. N-linked glycans: an underappreciated key determinant of T cell development, activation, and function. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00035. [PMID: 38027254 PMCID: PMC10662610 DOI: 10.1097/in9.0000000000000035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023]
Abstract
N-linked glycosylation is a post-translational modification that results in the decoration of newly synthesized proteins with diverse types of oligosaccharides that originate from the amide group of the amino acid asparagine. The sequential and collective action of multiple glycosidases and glycosyltransferases are responsible for determining the overall size, composition, and location of N-linked glycans that become covalently linked to an asparagine during and after protein translation. A growing body of evidence supports the critical role of N-linked glycan synthesis in regulating many features of T cell biology, including thymocyte development and tolerance, as well as T cell activation and differentiation. Here, we provide an overview of how specific glycosidases and glycosyltransferases contribute to the generation of different types of N-linked glycans and how these post-translational modifications ultimately regulate multiple facets of T cell biology.
Collapse
Affiliation(s)
- Mahmoud Abdelbary
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey C. Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
6
|
Kremsreiter SM, Kroell ASH, Weinberger K, Boehm H. Glycan-Lectin Interactions in Cancer and Viral Infections and How to Disrupt Them. Int J Mol Sci 2021; 22:10577. [PMID: 34638920 PMCID: PMC8508825 DOI: 10.3390/ijms221910577] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Glycan-lectin interactions play an essential role in different cellular processes. One of their main functions is involvement in the immune response to pathogens or inflammation. However, cancer cells and viruses have adapted to avail themselves of these interactions. By displaying specific glycosylation structures, they are able to bind to lectins, thus promoting pathogenesis. While glycan-lectin interactions promote tumor progression, metastasis, and/or chemoresistance in cancer, in viral infections they are important for viral entry, release, and/or immune escape. For several years now, a growing number of investigations have been devoted to clarifying the role of glycan-lectin interactions in cancer and viral infections. Various overviews have already summarized and highlighted their findings. In this review, we consider the interactions of the lectins MGL, DC-SIGN, selectins, and galectins in both cancer and viral infections together. A possible transfer of ways to target and disrupt them might lead to new therapeutic approaches in different pathological backgrounds.
Collapse
Affiliation(s)
- Stefanie Maria Kremsreiter
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Ruprecht Karls University Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany; (S.M.K.); (A.-S.H.K.); (K.W.)
| | - Ann-Sophie Helene Kroell
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Ruprecht Karls University Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany; (S.M.K.); (A.-S.H.K.); (K.W.)
| | - Katharina Weinberger
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Ruprecht Karls University Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany; (S.M.K.); (A.-S.H.K.); (K.W.)
| | - Heike Boehm
- Max-Planck-Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
| |
Collapse
|
7
|
He YS, Hu YQ, Xiang K, Chen Y, Feng YT, Yin KJ, Huang JX, Wang J, Wu ZD, Wang GH, Pan HF. Therapeutic potential of galectin-1 and galectin-3 in autoimmune diseases. Curr Pharm Des 2021; 28:36-45. [PMID: 34579628 DOI: 10.2174/1381612827666210927164935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/24/2021] [Indexed: 11/22/2022]
Abstract
Galectins are a highly conserved protein family that binds to β-galactosides. Different members of this family play a variety of biological functions in physiological and pathological processes such as angiogenesis, regulation of immune cell activity, and cell adhesion. Galectins are widely distributed and play a vital role both inside and outside cells. It can regulate homeostasis and immune function in vivo through mechanisms such as apoptosis. Recent studies indicate that galectins exhibit pleiotropic roles in inflammation. Furthermore, emerging studies have found that galectins are involved in the occurrence and development of autoimmune diseases such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), type 1 diabetes (T1D) and systemic sclerosis (SSc) by regulating cell adhesion, apoptosis, and other mechanisms. This review will briefly discuss the biological characteristics of the two most widely expressed and extensively explored members of the galectin family, galectin-1 and galectin-3, as well as their pathogenetic and therapeutic roles in autoimmune diseases. These information may provide a novel and promising therapeutic target for autoimmune diseases.
Collapse
Affiliation(s)
- Yi-Sheng He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui. China
| | - Yu-Qian Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui. China
| | - Kun Xiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui. China
| | - Yue Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui. China
| | - Ya-Ting Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui. China
| | - Kang-Jia Yin
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui. China
| | - Ji-Xiang Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui. China
| | - Jie Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui. China
| | - Zheng-Dong Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui. China
| | - Gui-Hong Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui. China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui. China
| |
Collapse
|
8
|
Unraveling How Tumor-Derived Galectins Contribute to Anti-Cancer Immunity Failure. Cancers (Basel) 2021; 13:cancers13184529. [PMID: 34572756 PMCID: PMC8469970 DOI: 10.3390/cancers13184529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/16/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary This review compiles our current knowledge of one of the main pathways activated by tumors to escape immune attack. Indeed, it integrates the current understanding of how tumor-derived circulating galectins affect the elicitation of effective anti-tumor immunity. It focuses on several relevant topics: which are the main galectins produced by tumors, how soluble galectins circulate throughout biological liquids (taking a body-settled gradient concentration into account), the conditions required for the galectins’ functions to be accomplished at the tumor and tumor-distant sites, and how the physicochemical properties of the microenvironment in each tissue determine their functions. These are no mere semantic definitions as they define which functions can be performed in said tissues instead. Finally, we discuss the promising future of galectins as targets in cancer immunotherapy and some outstanding questions in the field. Abstract Current data indicates that anti-tumor T cell-mediated immunity correlates with a better prognosis in cancer patients. However, it has widely been demonstrated that tumor cells negatively manage immune attack by activating several immune-suppressive mechanisms. It is, therefore, essential to fully understand how lymphocytes are activated in a tumor microenvironment and, above all, how to prevent these cells from becoming dysfunctional. Tumors produce galectins-1, -3, -7, -8, and -9 as one of the major molecular mechanisms to evade immune control of tumor development. These galectins impact different steps in the establishment of the anti-tumor immune responses. Here, we carry out a critical dissection on the mechanisms through which tumor-derived galectins can influence the production and the functionality of anti-tumor T lymphocytes. This knowledge may help us design more effective immunotherapies to treat human cancers.
Collapse
|
9
|
Dings RPM, Kumar N, Mikkelson S, Gabius HJ, Mayo KH. Simulating cellular galectin networks by mixing galectins in vitro reveals synergistic activity. Biochem Biophys Rep 2021; 28:101116. [PMID: 34485713 PMCID: PMC8408429 DOI: 10.1016/j.bbrep.2021.101116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/09/2021] [Accepted: 08/23/2021] [Indexed: 12/31/2022] Open
Abstract
Background Even though members of the family of adhesion/growth-regulatory galectins are increasingly detected to be co-expressed, they are still being routinely tested separately. The recent discovery of heterodimer formation among galectins-1, -3, and -7 in mixtures prompts further study of their functional activities in mixtures. Methods Cell agglutination, galectin binding to cells, as well as effects on cell proliferation, onset of apoptosis and migration were determined in assays using various cell types and mixtures of galectins-1, -3, and -7. Results Evidence for a more than additive increases of experimental parameters was consistently obtained. Conclusion Testing galectins in mixtures simulates the situation of co-expression in situ and reveals unsuspected over-additive activities. This new insight is relevant for analyzing galectin functionality in (patho)physiological conditions. Galectins-1, -3, and -7 form heterodimers in solution. Mixtures of galectins simulates galectin co-expression in situ. Mixtures display synergistic activities in vitro. Cell agglutination, apoptosis, proliferation, migration affected. Findings are relevant for galectin functionality in vivo.
Collapse
Affiliation(s)
- Ruud P M Dings
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Nigam Kumar
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Sterling Mikkelson
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Hans-Joachim Gabius
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University, Veterinarstr. 13, Munich, 80539, Germany
| | - Kevin H Mayo
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
10
|
The Role of Glycosylation in Inflammatory Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1325:265-283. [PMID: 34495540 DOI: 10.1007/978-3-030-70115-4_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The diversity of glycan presentation in a cell, tissue and organism is enormous, which reflects the huge amount of important biological information encoded by the glycome which has not been fully understood. A compelling body of evidence has been highlighting the fundamental role of glycans in immunity, such as in development, and in major inflammatory processes such as inflammatory bowel disease, systemic lupus erythematosus and other autoimmune disorders. Glycans play an instrumental role in the immune response, integrating the canonical circuits that regulate innate and adaptive immune responses. The relevance of glycosylation in immunity is demonstrated by the role of glycans as important danger-associated molecular patterns and pathogen-associated molecular patterns associated with the discrimination between self and non-self; also as important regulators of the threshold of T cell activation, modulating receptors signalling and the activity of both T and other immune cells. In addition, glycans are important determinants that regulate the dynamic crosstalk between the microbiome and immune response. In this chapter, the essential role of glycans in the immunopathogenesis of inflammatory disorders will be presented and its potential clinical applications (diagnosis, prognosis and therapeutics) will be highlighted.
Collapse
|
11
|
Manero-Rupérez N, Martínez-Bosch N, Barranco LE, Visa L, Navarro P. The Galectin Family as Molecular Targets: Hopes for Defeating Pancreatic Cancer. Cells 2020; 9:E689. [PMID: 32168866 PMCID: PMC7140611 DOI: 10.3390/cells9030689] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/21/2020] [Accepted: 02/27/2020] [Indexed: 12/14/2022] Open
Abstract
Galectins are a family of proteins that bind β-galactose residues through a highly conserved carbohydrate recognition domain. They regulate several important biological functions, including cell proliferation, adhesion, migration, and invasion, and play critical roles during embryonic development and cell differentiation. In adults, different galectin members are expressed depending on the tissue type and can be altered during pathological processes. Numerous reports have shown the involvement of galectins in diseases, mostly inflammation and cancer. Here, we review the state-of-the-art of the role that different galectin family members play in pancreatic cancer. This tumor is predicted to become the second leading cause of cancer-related deaths in the next decade as there is still no effective treatment nor accurate diagnosis for it. We also discuss the possible translation of recent results about galectin expression and functions in pancreatic cancer into clinical interventions (i.e., diagnosis, prediction of prognosis and/or therapy) for this fatal disease.
Collapse
Affiliation(s)
- Noemí Manero-Rupérez
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain;
| | - Neus Martínez-Bosch
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain;
| | - Luis E Barranco
- Department of Gastroenterolgy, Hospital del Mar-IMIM, 08003 Barcelona, Spain;
| | - Laura Visa
- Department of Medical Oncology, Hospital del Mar-IMIM-CIBERONC, 08003 Barcelona, Spain;
| | - Pilar Navarro
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain
- Institute of Biomedical Research of Barcelona (IIBB-CSIC), 08036 Barcelona, Spain
- Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
| |
Collapse
|
12
|
Castro LL, Kitoko JZ, Xisto DG, Olsen PC, Guedes HLM, Morales MM, Lopes-Pacheco M, Cruz FF, Rocco PRM. Multiple doses of adipose tissue-derived mesenchymal stromal cells induce immunosuppression in experimental asthma. Stem Cells Transl Med 2019; 9:250-260. [PMID: 31746562 PMCID: PMC6988761 DOI: 10.1002/sctm.19-0120] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 10/09/2019] [Indexed: 12/14/2022] Open
Abstract
In experimental house dust mite (HDM)‐induced allergic asthma, therapeutic administration of a single dose of adipose tissue‐derived mesenchymal stromal cells (MSCs) ameliorates lung inflammation but is unable to reverse remodeling. We hypothesized that multiple doses of MSCs might exert better therapeutic effects by reducing lung inflammation and remodeling but might also result in immunosuppressive effects in experimental asthma. HDM was administered intranasally in C57BL/6 mice. After the last HDM challenge, mice received two or three doses of MSCs (105 cells per day) or saline intravenously. An additional cohort of mice received dexamethasone as a positive control for immunosuppression. Two and three doses of MSCs reduced lung inflammation, levels of interleukin (IL)‐4, IL‐13, and eotaxin; total leukocyte, CD4+ T‐cell, and eosinophil counts in bronchoalveolar lavage fluid; and total leukocyte counts in bone marrow, spleen, and mediastinal lymph nodes. Two and three doses of MSCs also reduced collagen fiber content and transforming growth factor‐β levels in lung tissue; however, the three‐dose regimen was more effective, and reduced these parameters to control levels, while also decreasing α‐actin content in lung tissue. Two and three doses of MSCs improved lung mechanics. Dexamethasone, two and three doses of MSCs similarly increased galectin levels, but only the three‐dose regimen increased CD39 levels in the thymus. Dexamethasone and the three‐dose, but not the two‐dose regimen, also increased levels of programmed death receptor‐1 and IL‐10, while reducing CD4+CD8low cell percentage in the thymus. In conclusion, multiple doses of MSCs reduced lung inflammation and remodeling while causing immunosuppression in HDM‐induced allergic asthma.
Collapse
Affiliation(s)
- Ligia L Castro
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Jamil Z Kitoko
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Clinical Bacteriology and Immunology, School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Debora G Xisto
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscilla C Olsen
- Laboratory of Clinical Bacteriology and Immunology, School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Herbert L M Guedes
- Laboratory of Glycobiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo M Morales
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.,Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.,Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Fettis MM, Farhadi SA, Hudalla GA. A chimeric, multivalent assembly of galectin-1 and galectin-3 with enhanced extracellular activity. Biomater Sci 2019; 7:1852-1862. [PMID: 30899922 DOI: 10.1039/c8bm01631c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Galectins are attractive therapeutic candidates to control aberrant immune system activation because they can alter the phenotype and function of various innate and adaptive immune cells. However, use of exogenous galectin-1 ("G1") and galectin-3 ("G3") as immunomodulators is challenged by their high dosing requirements and dynamic quaternary structures. Here we report a chimeric assembly of G1 and G3 with enhanced extracellular activity ("G1/G3 Zipper"), which was created by recombinant fusion of G1 and G3 via a peptide linker that forms a two-stranded α-helical coiled-coil. G1/G3 Zipper had higher apparent binding affinity for immobilized lactose and a lower concentration threshold for inducing soluble glycoprotein crosslinking than G1, a recombinant fusion of G1 and G3 with a flexible peptide linker ("G1/G3"), or a recently reported stable G1 dimer crosslinked by poly(ethylene glycol) diacrylate ("G1-PEG-G1"). As a result, G1/G3 Zipper was more effective at inducing Jurkat T cell apoptosis in media containing serum, and was the only variant that could induce apoptosis at low concentrations under serum-free conditions. The monomeric G1/G3 fusion protein lacked extracellular activity under all conditions tested, suggesting that the enhanced activity of G1/G3 Zipper was due to its quaternary structure and increased carbohydrate-recognition domain valency. Thus, combining G1 and G3 into a non-native chimeric assembly provides a new candidate therapeutic with greater immunomodulatory potency than the wild-type proteins and previously reported engineered variants.
Collapse
Affiliation(s)
- Margaret M Fettis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA 32611.
| | | | | |
Collapse
|
14
|
Abel WF, Funk CR, Blenda AV. Galectins in the Pathogenesis of Cerebrovascular Accidents: An Overview. J Exp Neurosci 2019; 13:1179069519836794. [PMID: 31007530 PMCID: PMC6458655 DOI: 10.1177/1179069519836794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/19/2019] [Indexed: 01/04/2023] Open
Abstract
Due to limitations of neuroimaging, such as the isodense appearance of blood to neuronal tissue in subacute hemorrhagic stroke, a body of studies have been performed to evaluate candidate biomarkers which may aid in accurate determination of cerebrovascular accident type. Beyond aiding in the delineation of stroke cause, biomarkers could also confer useful prognostic information to help clinicians plan use of resources. One of the candidate biomarkers studied for detection of cerebrovascular accident (CVA) includes a class of proteins called galectins. Galectins bind β-galactoside through a highly conserved carbohydrate recognition domain, endowing an ability to interact with carbohydrate moieties on glycoproteins, some of which are relevant to CVA response. Furthermore, galectins-1, -2, -3, -9, and -12 are expressed in tissues relevant to CVA, and some exhibit characteristics (eg, extracellular secretion) that could render feasible their detection in serum. Galectins-1 and -3 appear to have the largest amounts of preclinical evidence, consistently demonstrating increased activity and expression levels during CVA. However, a lack of standardization of biochemical assays across cohort studies limits further translation of these basic science studies. This review aims to increase awareness of the biochemical roles of galectins in CVA, while also highlighting challenges and remaining questions preventing the translation of basic science observations into a clinically useful test.
Collapse
Affiliation(s)
- William F Abel
- University of South Carolina School of Medicine Greenville, Greenville, SC, USA
| | | | - Anna V Blenda
- University of South Carolina School of Medicine Greenville, Greenville, SC, USA
| |
Collapse
|
15
|
Tsutsui S, Yoshinaga T, Watanabe S, Tsukamoto K, Nakamura O. Mucosal galectin genes in all freshwater eels of the genus Anguilla. JOURNAL OF FISH BIOLOGY 2019; 94:660-670. [PMID: 30779133 DOI: 10.1111/jfb.13936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/18/2019] [Indexed: 06/09/2023]
Abstract
In this study, we determined the genomic DNA sequences of the mucosal galectin-encoding genes from all 19 species and subspecies of the genus Anguilla. The nucleotide sequences of the galectin genes were c. 2.3-2.5 kb long and the organisation of their four exons and three introns was conserved in all species. An unusual sequence was found in the fourth exon of Anguilla reinhardtii, resulting in a unique deduced amino-acid sequence at the C-terminus. All six amino-acid residues important for β-galactoside binding were conserved in three species, while one residue (R73 ) was substituted to K73 in the other 16 species-subspecies, including Anguilla marmorata. However, this substitution did not appear to affect the sugar-binding ability of galectins because the galectin of A. marmorata was previously shown to bind to lactose. We also discuss the molecular evolution of galectins among Anguilla spp. and the homologues previously identified in Conger myriaster.
Collapse
Affiliation(s)
- Shigeyuki Tsutsui
- School of Marine Biosciences, Kitasato University, 1-15-1, Kitasato, Sagamihara, Kanagawa 252-0373, Japan
| | - Tatsuki Yoshinaga
- School of Marine Biosciences, Kitasato University, 1-15-1, Kitasato, Sagamihara, Kanagawa 252-0373, Japan
| | - Shun Watanabe
- Faculty of Agriculture, Kindai University, Nara 631-0052, Japan
| | - Katsumi Tsukamoto
- College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa 252-0880, Japan
| | - Osamu Nakamura
- School of Marine Biosciences, Kitasato University, 1-15-1, Kitasato, Sagamihara, Kanagawa 252-0373, Japan
| |
Collapse
|
16
|
Riess JW, Kong CS, West RB, Padda SK, Neal JW, Wakelee HA, Le QT. Increased Galectin-1 Expression in Thymic Epithelial Tumors. Clin Lung Cancer 2019; 20:e356-e361. [PMID: 30773448 DOI: 10.1016/j.cllc.2018.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/25/2018] [Accepted: 12/08/2018] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Thymic epithelial tumors (TET) are rare malignancies with a paucity of data on biology and therapeutics. Galectin-1 is a member of the β-galactoside binding protein family and has been shown to mediate tumor growth via modulation of immune cell function. This study examined galectin-1 expression in TET. PATIENTS AND METHODS A tissue microarray of 68 patients with TET and 8 benign thymus controls were stained for galectin-1 expression and scored by a pathologist blinded to patient clinical and pathologic data. Galectin-1 expression +1 or greater staining intensity was considered positive. Clinical and pathologic data were abstracted from institutional databases. Expression of galectin-1 in thymic tumor was compared to benign thymus controls and correlated with pertinent clinical and pathologic data. RESULTS Galectin-1 expression was higher in TET compared to benign thymus controls (65% vs. 0%). No significant association between galectin-1 expression and the development of recurrent disease, paraneoplastic syndromes, or overall survival was noted. CONCLUSION Galectin-1 is overexpressed in the majority of TET. Detection of galectin-1 may differentiate benign from neoplastic thymic processes. Additional studies are needed to assess the role of galectin-1 in the development of TET.
Collapse
Affiliation(s)
- Jonathan W Riess
- Division of Hematology/Oncology, Department of Internal Medicine, UC Davis School of Medicine, UC Davis Comprehensive Cancer Center, Sacramento, CA.
| | - Christina S Kong
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Robert B West
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Sukhmani K Padda
- Division of Oncology, Department of Medicine, Stanford University School of Medicine and Stanford Cancer Institute, Stanford, CA
| | - Joel W Neal
- Division of Oncology, Department of Medicine, Stanford University School of Medicine and Stanford Cancer Institute, Stanford, CA
| | - Heather A Wakelee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine and Stanford Cancer Institute, Stanford, CA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine Stanford Cancer Institute, Stanford, CA
| |
Collapse
|
17
|
Giovannone N, Smith LK, Treanor B, Dimitroff CJ. Galectin-Glycan Interactions as Regulators of B Cell Immunity. Front Immunol 2018; 9:2839. [PMID: 30564237 PMCID: PMC6288978 DOI: 10.3389/fimmu.2018.02839] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Cell surface glycans and their glycan-binding partners (lectins) have generally been recognized as adhesive assemblies with neighbor cells or matrix scaffolds in organs and the blood stream. However, our understanding of the roles for glycan-lectin interactions in immunity has expanded substantially to include regulation of nearly every stage of an immune response, from pathogen sensing to immune contraction. In this Mini-Review, we discuss the role of the ß-galactoside-binding lectins known as galectins specifically in the regulation of B-lymphocyte (B cell) development, activation, and differentiation. In particular, we highlight several recent studies revealing new roles for galectin (Gal)-9 in the modulation of B cell receptor-mediated signaling and activation in mouse and man. The roles for cell surface glycosylation, especially I-branching of N-glycans synthesized by the glycosyltransferase GCNT2, in the regulation of Gal-9 binding activity are also detailed. Finally, we consider how dysregulation of these factors may contribute to aberrant immune activation and autoimmune disease.
Collapse
Affiliation(s)
- Nicholas Giovannone
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Logan K. Smith
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Bebhinn Treanor
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada
| | - Charles J. Dimitroff
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| |
Collapse
|
18
|
Nielsen MI, Stegmayr J, Grant OC, Yang Z, Nilsson UJ, Boos I, Carlsson MC, Woods RJ, Unverzagt C, Leffler H, Wandall HH. Galectin binding to cells and glycoproteins with genetically modified glycosylation reveals galectin-glycan specificities in a natural context. J Biol Chem 2018; 293:20249-20262. [PMID: 30385505 DOI: 10.1074/jbc.ra118.004636] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/22/2018] [Indexed: 01/02/2023] Open
Abstract
Galectins compose a protein family defined by a conserved sequence motif conferring affinity for β-galactose-containing glycans. Moreover, galectins gain higher affinity and fine-tune specificity by glycan interactions at sites adjacent to their β-galactoside-binding site, as revealed by extensive testing against panels of purified glycans. However, in cells, galectins bind glycans on glycoproteins and glycolipids in the context of other cellular components, such as at the cell surface. Because of difficulties in characterizing natural cellular environments, we currently lack a detailed understanding of galectin-binding specificities in the cellular context. To address this challenge, we used a panel of genetically stable glycosylation mutated CHO cells that express defined glycans to evaluate the binding affinities of 10 different carbohydrate-recognition domains in galectins to N-glycans and mucin-type O-glycans. Using flow cytometry, we measured the cell-surface binding of the galectins. Moreover, we used fluorescence anisotropy to determine the galectin affinities to recombinant erythropoietin used as a reporter glycoprotein produced by the glycoengineered cells and to synthetic N-glycans with defined branch structures. We found that all galectins, apart from galectin-8N, require complex N-glycans for high-affinity binding. Galectin-8N targeted both N- and O-linked glycans with high affinity, preferring 2,3-sialylated N-acetyllactosamine (LacNAc) structures. Furthermore, we found that 2,3-sialylation suppresses high-affinity binding of select galectins, including galectin-2, -3, -4N, and -7. Structural modeling provided a basis for interpreting the observed binding preferences. These results underscore the power of a glycoengineered platform to dissect the glycan-binding specificities of carbohydrate-binding proteins.
Collapse
Affiliation(s)
- Mathias Ingemann Nielsen
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - John Stegmayr
- the Division for Microbiology, Immunology and Glycobiology (MIG), Department of Laboratory Medicine, Faculty of Medicine, Lund University, 22100 Lund, Sweden
| | - Oliver C Grant
- the Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | - Zhang Yang
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Ulf J Nilsson
- the Centre for Analysis and Synthesis, Department of Chemistry, Lund University, 22100 Lund, Sweden, and
| | - Irene Boos
- the Bioorganische Chemie, Universität Bayreuth, Gebäude NW I, 95440 Bayreuth, Germany
| | - Michael C Carlsson
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark,; the Division for Microbiology, Immunology and Glycobiology (MIG), Department of Laboratory Medicine, Faculty of Medicine, Lund University, 22100 Lund, Sweden
| | - Robert J Woods
- the Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602
| | - Carlo Unverzagt
- the Bioorganische Chemie, Universität Bayreuth, Gebäude NW I, 95440 Bayreuth, Germany
| | - Hakon Leffler
- the Division for Microbiology, Immunology and Glycobiology (MIG), Department of Laboratory Medicine, Faculty of Medicine, Lund University, 22100 Lund, Sweden
| | - Hans H Wandall
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark,.
| |
Collapse
|
19
|
Zhao T, Wei X, Yang J, Wang S, Zhang Y. Galactoside-binding lectin in Solen grandis as a pattern recognition receptor mediating opsonization. FISH & SHELLFISH IMMUNOLOGY 2018; 82:183-189. [PMID: 30107261 DOI: 10.1016/j.fsi.2018.08.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/31/2018] [Accepted: 08/11/2018] [Indexed: 06/08/2023]
Abstract
Galactoside-binding lectin (galectin) is a type of pathogen recognition molecule that occupies an important position in the invertebrate innate immunity system. Our previous study has identified a galectin gene in mollusk Solen grandis (SgGal-1) and illustrated its potential roles in innate immunity. By the functional study using recombinant protein and specific antibody, here, we confirmed the pivotal roles of SgGal-1 in immune defense of S. grandis. SgGal-1 protein was expressed in many tested tissues including gill, mantle, hepatopancreas and gonad, except hemocytes and muscle. The recombinant SgGal-1 (rSgGal-1) bound PGN and β-glucan instead of LPS in vitro, and it further caused significant agglutination of five different microbes, suggesting SgGal-1 served as a pattern recognition receptor (PRR) involved in immune defense of mollusk. Furthermore, SgGal-1 recruited hemocytes to encapsulate, which was blocked by anti-rSgGal-1 serum. In the meantime, rSgGal-1 as well as promoted the phagocytosis of hemocytes against Escherichia coli in vitro. All these results suggested that SgGal-1 in S. grandis not only acted as a PRR recognizing microbes but also directly participated in the process of immune opsonization to protect the host from pathogenic infection.
Collapse
Affiliation(s)
- Tianyu Zhao
- Laboratory of Aquatic Comparative Immunology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiumei Wei
- Laboratory of Aquatic Comparative Immunology, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Jialong Yang
- Laboratory of Aquatic Comparative Immunology, School of Life Sciences, East China Normal University, Shanghai 200241, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Sheng Wang
- Shandong Marine Resource and Environment Research Institute, Yantai 264006, China
| | - Yu Zhang
- Laboratory of Aquatic Comparative Immunology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
20
|
Cameron G, Godfrey DI. Differential surface phenotype and context-dependent reactivity of functionally diverse NKT cells. Immunol Cell Biol 2018; 96:759-771. [PMID: 29504657 DOI: 10.1111/imcb.12034] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/20/2018] [Accepted: 02/28/2018] [Indexed: 12/28/2022]
Abstract
Natural Killer T (NKT) cells are a functionally diverse population that recognizes lipid-based antigens in association with the antigen-presenting molecule CD1d. Here, we define a technique to separate the functionally distinct thymic NKT1, NKT2 and NKT17 cell subsets by their surface expression of CD278 (ICOS) and the activation-associated glycoform of CD43, enabling the investigation of subset-specific effector-functions. We report that all three subsets express the transcription factor GATA-3 and the potential to produce IL-4 and IL-10 following activation. This questions the notion that NKT2 cells are the predominant source of IL-4 within the NKT cell pool, and suggests that IL-10-production may be more indicative of NKT cell plasticity than the existence of a distinct regulatory lineage or subset. We also show that many NKT17 cells are CD4+ and are biased toward Vβ8.3 TCR gene usage. Lastly, we demonstrate that the toll-like receptor (TLR) ligand lipopolysaccharide (LPS) can induce a NKT17 cell-biased response, even in the absence of exogenous antigen, and that combining LPS with α-GalCer resulted in enhanced IL-17A-production, and reduced levels of the immunosuppressive cytokine IL-10. This study provides a novel means to examine the context-dependent reactivity of the functionally heterogeneous NKT cell population and provides important new insight into the functional biology of these subsets.
Collapse
Affiliation(s)
- Garth Cameron
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, 3000, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, 3000, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
21
|
Micheau O. Regulation of TNF-Related Apoptosis-Inducing Ligand Signaling by Glycosylation. Int J Mol Sci 2018; 19:E715. [PMID: 29498673 PMCID: PMC5877576 DOI: 10.3390/ijms19030715] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/19/2018] [Accepted: 02/24/2018] [Indexed: 12/26/2022] Open
Abstract
Tumor necrosis-factor related apoptosis-inducing ligand, also known as TRAIL or APO2L (Apo-2 ligand), is a cytokine of the TNF superfamily acknowledged for its ability to trigger selective apoptosis in tumor cells while being relatively safe towards normal cells. Its binding to its cognate agonist receptors, namely death receptor 4 (DR4) and/or DR5, can induce the formation of a membrane-bound macromolecular complex, coined DISC (death-signaling inducing complex), necessary and sufficient to engage the apoptotic machinery. At the very proximal level, TRAIL DISC formation and activation of apoptosis is regulated both by antagonist receptors and by glycosylation. Remarkably, though, despite the fact that all membrane-bound TRAIL receptors harbor putative glycosylation sites, only pro-apoptotic signaling through DR4 and DR5 has, so far, been found to be regulated by N- and O-glycosylation, respectively. Because putative N-glycosylation sequons and O-glycosylation sites are also found and conserved in all these receptors throughout all animal species (in which these receptors have been identified), glycosylation is likely to play a more prominent role than anticipated in regulating receptor/receptor interactions or trafficking, ultimately defining cell fate through TRAIL stimulation. This review aims to present and discuss these emerging concepts, the comprehension of which is likely to lead to innovative anticancer therapies.
Collapse
Affiliation(s)
- Olivier Micheau
- INSERM, UMR1231, Laboratoire d'Excellence LipSTIC, F-21079 Dijon, France.
- UFR Sciences de Santé, University Bourgogne Franche-Comté, UBFC, F-21079 Dijon, France.
| |
Collapse
|
22
|
Acar S, Paketçi A, Küme T, Tuhan H, Gürsoy Çalan Ö, Demir K, Böber E, Abacı A. Serum galectin-1 levels are positively correlated with body fat and negatively with fasting glucose in obese children. Peptides 2017; 95:51-56. [PMID: 28728946 DOI: 10.1016/j.peptides.2017.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/08/2017] [Accepted: 07/13/2017] [Indexed: 01/03/2023]
Abstract
Galectin-1, a recently identified peptide, is primarily released from the adipose tissue. Although galectin-1 was shown to have an anti-inflammatory effect, its specific function is not clearly understood. We aimed to evaluate the relationship of serum galectin-1 levels with clinical and laboratory parameters in childhood obesity. A total of 45 obese children (mean age: 12.1±3.1years) and 35 normal-weight children (mean age: 11.8±2.2years) were enrolled. Clinical [body mass index (BMI), waist circumference (WC), percentage of body fat and blood pressure] and biochemical [glucose, insulin, lipids, galectin-1, high-sensitive C-reactive protein (hsCRP) and leptin levels] parameters were assessed. Serum galectin-1, hsCRP and leptin levels were significantly higher in obese children than those in normal-weight children (12.4 vs 10.2ng/mL, p<0.001; 3.28 vs 0.63mg/L, p<0.001; 8.3 vs 1.2ng/mL, p<0.001, respectively). In obese children, galectin-1 levels correlated negatively with fasting glucose (r=-0.346, p=0.020) and positively with fat mass (r=0.326, p=0.026) and WC standard deviation score (SDS) (r=0.451, p=0.002). The multivariate regression analysis demonstrated that serum galectin-1 levels were significantly associated with fasting glucose and WC SDS. This study showed that obese children had significantly higher galectin-1 levels in proportion to fat mass in obese cases than those in healthy children, which may be interpreted as a compensatory increase in an attempt to improve glucose metabolism.
Collapse
Affiliation(s)
- Sezer Acar
- Division of Pediatric Endocrinology, Dokuz Eylul University School of Medicine, Izmir, Turkey
| | - Ahu Paketçi
- Division of Pediatric Endocrinology, Dokuz Eylul University School of Medicine, Izmir, Turkey
| | - Tuncay Küme
- Department of Biochemistry, Dokuz Eylul University School of Medicine, Izmir, Turkey
| | - Hale Tuhan
- Division of Pediatric Endocrinology, Dokuz Eylul University School of Medicine, Izmir, Turkey
| | - Özlem Gürsoy Çalan
- Department of Biochemistry, Dokuz Eylul University School of Medicine, Izmir, Turkey
| | - Korcan Demir
- Division of Pediatric Endocrinology, Dokuz Eylul University School of Medicine, Izmir, Turkey
| | - Ece Böber
- Division of Pediatric Endocrinology, Dokuz Eylul University School of Medicine, Izmir, Turkey
| | - Ayhan Abacı
- Division of Pediatric Endocrinology, Dokuz Eylul University School of Medicine, Izmir, Turkey.
| |
Collapse
|
23
|
Abstract
Polymorphonuclear neutrophils (PMNs) are innate immune system cells that play an essential role in eradicating invading pathogens. PMN migration to sites of infection/inflammation requires exiting the microcirculation and subsequent crossing of epithelial barriers in mucosa-lined organs such as the lungs and intestines. Although these processes usually occur without significant damage to surrounding host tissues, dysregulated/excessive PMN transmigration and resultant bystander-tissue damage are characteristic of numerous mucosal inflammatory disorders. Mechanisms controlling PMN extravasation have been well characterized, but the molecular details regarding regulation of PMN migration across mucosal epithelia are poorly understood. Given that PMN migration across mucosal epithelia is strongly correlated with disease symptoms in many inflammatory mucosal disorders, enhanced understanding of the mechanisms regulating PMN transepithelial migration should provide insights into clinically relevant tissue-targeted therapies aimed at ameliorating PMN-mediated bystander-tissue damage. This review will highlight current understanding of the molecular interactions between PMNs and mucosal epithelia and the associated functional consequences.
Collapse
Affiliation(s)
- Jennifer C Brazil
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
24
|
Wu X, Li J, Connolly EM, Liao X, Ouyang J, Giobbie-Hurder A, Lawrence D, McDermott D, Murphy G, Zhou J, Piesche M, Dranoff G, Rodig S, Shipp M, Hodi FS. Combined Anti-VEGF and Anti-CTLA-4 Therapy Elicits Humoral Immunity to Galectin-1 Which Is Associated with Favorable Clinical Outcomes. Cancer Immunol Res 2017; 5:446-454. [PMID: 28473314 DOI: 10.1158/2326-6066.cir-16-0385] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/03/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023]
Abstract
The combination of anti-VEGF blockade (bevacizumab) with immune checkpoint anti-CTLA-4 blockade (ipilimumab) in a phase I study showed tumor endothelial activation and immune cell infiltration that were associated with favorable clinical outcomes in patients with metastatic melanoma. To identify potential immune targets responsible for these observations, posttreatment plasma from long-term responding patients were used to screen human protein arrays. We reported that ipilimumab plus bevacizumab therapy elicited humoral immune responses to galectin-1 (Gal-1), which exhibits protumor, proangiogenesis, and immunosuppressive activities in 37.2% of treated patients. Gal-1 antibodies purified from posttreatment plasma suppressed the binding of Gal-1 to CD45, a T-cell surface receptor that transduces apoptotic signals upon binding to extracellular Gal-1. Antibody responses to Gal-1 were found more frequently in the group of patients with therapeutic responses and correlated with improved overall survival. In contrast, another subgroup of treated patients had increased circulating Gal-1 protein instead, and they had reduced overall survival. Our findings suggest that humoral immunity to Gal-1 may contribute to the efficacy of anti-VEGF and anti-CTLA-4 combination therapy. Gal-1 may offer an additional therapeutic target linking anti-angiogenesis and immune checkpoint blockade. Cancer Immunol Res; 5(6); 446-54. ©2017 AACR.
Collapse
Affiliation(s)
- Xinqi Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Melanoma Disease Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Center for Immuno-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Jingjing Li
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Melanoma Disease Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Center for Immuno-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Erin M Connolly
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Melanoma Disease Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Center for Immuno-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Xiaoyun Liao
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Melanoma Disease Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Center for Immuno-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Jing Ouyang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Anita Giobbie-Hurder
- Department of Biostatistics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Donald Lawrence
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | | | - George Murphy
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jun Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Melanoma Disease Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Center for Immuno-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Matthias Piesche
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | - Glenn Dranoff
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Scott Rodig
- Center for Immuno-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Margaret Shipp
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Center for Immuno-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts. .,Melanoma Disease Center, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts.,Center for Immuno-Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
25
|
Coelho LCBB, Silva PMDS, Lima VLDM, Pontual EV, Paiva PMG, Napoleão TH, Correia MTDS. Lectins, Interconnecting Proteins with Biotechnological/Pharmacological and Therapeutic Applications. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2017; 2017:1594074. [PMID: 28367220 PMCID: PMC5359455 DOI: 10.1155/2017/1594074] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 01/21/2017] [Accepted: 02/06/2017] [Indexed: 11/18/2022]
Abstract
Lectins are proteins extensively used in biomedical applications with property to recognize carbohydrates through carbohydrate-binding sites, which identify glycans attached to cell surfaces, glycoconjugates, or free sugars, detecting abnormal cells and biomarkers related to diseases. These lectin abilities promoted interesting results in experimental treatments of immunological diseases, wounds, and cancer. Lectins obtained from virus, microorganisms, algae, animals, and plants were reported as modulators and tool markers in vivo and in vitro; these molecules also play a role in the induction of mitosis and immune responses, contributing for resolution of infections and inflammations. Lectins revealed healing effect through induction of reepithelialization and cicatrization of wounds. Some lectins have been efficient agents against virus, fungi, bacteria, and helminths at low concentrations. Lectin-mediated bioadhesion has been an interesting characteristic for development of drug delivery systems. Lectin histochemistry and lectin-based biosensors are useful to detect transformed tissues and biomarkers related to disease occurrence; antitumor lectins reported are promising for cancer therapy. Here, we address lectins from distinct sources with some biological effect and biotechnological potential in the diagnosis and therapeutic of diseases, highlighting many advances in this growing field.
Collapse
Affiliation(s)
| | - Priscila Marcelino dos Santos Silva
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego 1235, Cidade Universitária, 50.670-901 Recife, PE, Brazil
| | - Vera Lúcia de Menezes Lima
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego 1235, Cidade Universitária, 50.670-901 Recife, PE, Brazil
| | - Emmanuel Viana Pontual
- Departamento de Morfologia e Fisiologia Animal, Universidade Federal Rural de Pernambuco, Rua Dom Manuel de Medeiros, s/n, Dois Irmãos, 52171-900 Recife, PE, Brazil
| | - Patrícia Maria Guedes Paiva
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego 1235, Cidade Universitária, 50.670-901 Recife, PE, Brazil
| | - Thiago Henrique Napoleão
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego 1235, Cidade Universitária, 50.670-901 Recife, PE, Brazil
| | - Maria Tereza dos Santos Correia
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Av. Prof. Moraes Rego 1235, Cidade Universitária, 50.670-901 Recife, PE, Brazil
| |
Collapse
|
26
|
Hornung Á, Monostori É, Kovács L. Systemic lupus erythematosus in the light of the regulatory effects of galectin-1 on T-cell function. Lupus 2017; 26:339-347. [PMID: 28100106 DOI: 10.1177/0961203316686846] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Galectin-1 is an endogenous immunoregulatory lectin-type protein. Its most important effects are the inhibition of the differentiation and cytokine production of Th1 and Th17 cells, and the induction of apoptosis of activated T-cells. Galectin-1 has been identified as a key molecule in antitumor immune surveillance, and data are accumulating about the pathogenic role of its deficiency, and the beneficial effects of its administration in various autoimmune disease models. Initial animal and human studies strongly suggest deficiencies in both galectin-1 production and responsiveness in systemic lupus erythematosus (SLE) T-cells. Since lupus features widespread abnormalities in T-cell activation, differentiation and viability, in this review the authors wished to highlight potential points in T-cell signalling processes that may be influenced by galectin-1. These points include GM-1 ganglioside-mediated lipid raft aggregation, early activation signalling steps involving p56Lck, the exchange of the CD3 ζ-ZAP-70 to the FcRγ-Syk pathway, defective mitogen-activated protein kinase pathway activation, impaired regulatory T-cell function, the failure to suppress the activity of interleukin 17 (IL-17) producing T-cells, and decreased suppression of the PI3K-mTOR pathway by phosphatase and tensin homolog (PTEN). These findings place galectin-1 into the group of potential pathogenic molecules in SLE.
Collapse
Affiliation(s)
- Á Hornung
- 1 Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,2 Department of Rheumatology and Immunology, University of Szeged, Faculty of Medicine, Albert Szent-Györgyi Health Centre, Szeged, Hungary
| | - É Monostori
- 1 Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - L Kovács
- 2 Department of Rheumatology and Immunology, University of Szeged, Faculty of Medicine, Albert Szent-Györgyi Health Centre, Szeged, Hungary
| |
Collapse
|
27
|
Sasaguri K, Yamada K, Narimatsu Y, Oonuki M, Oishi A, Koda K, Kubo KY, Yamamoto T, Kadoya T. Stress-induced galectin-1 influences immune tolerance in the spleen and thymus by modulating CD45 immunoreactive lymphocytes. J Physiol Sci 2016; 67:489-496. [PMID: 27573167 PMCID: PMC5487876 DOI: 10.1007/s12576-016-0478-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 08/15/2016] [Indexed: 02/01/2023]
Abstract
Galectin-1 (Gal-1) is differentially expressed in normal and pathological tissues and regulates immune cell homeostasis. Restraint stress increases serum Gal-1 in rats. However, the function of stress-induced Gal-1 in serum is unknown. We determined if stress-induced Gal-1 in serum accumulates in immunocompetent organs as protection from physiological and/or psychological stress. Western blotting showed that the intensity of Gal-1 bands in stressed groups was significantly higher than that in controls. RT-PCR analysis indicated that the Gal-1 mRNA level did not increase after restraint stress. The numbers of Gal-1 immunoreactive cells in the splenic periarterial lymphatic sheath (PLS) and the thymus medulla of the stressed group were increased compared with those in controls. Furthermore, stress-induced Gal-1 immunoreactive cells corresponded to CD45 immunoreactive lymphocytes (CD45+) in the PLS of the spleen and the medulla of the thymus. Thus, stress-induced Gal-1 immediately accumulates in the spleen and thymus, and may modulate the immune response through apoptosis by binding to CD45+ lymphocytes in immune organs following physiological and/or psychological stress.
Collapse
Affiliation(s)
- Kenichi Sasaguri
- Department of Dentistry, Oral and Maxillofacial Surgery, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| | - Kentaro Yamada
- Brain Functions and Neuroscience Division, Department of Oral Science, Kanagawa Dental University Graduate School, Inaoka-cho 82, Yokosuka, Kanagawa, 238-0003, Japan
| | - Yuri Narimatsu
- Division of Orthodontics, Department of Oral Science, Kanagawa Dental University Graduate School, 82 Inaoka-cho, Yokosuka, Kanagawa, 238-8580, Japan
| | - Masami Oonuki
- Division of Orthodontics, Department of Oral Science, Kanagawa Dental University Graduate School, 82 Inaoka-cho, Yokosuka, Kanagawa, 238-8580, Japan
| | - Azusa Oishi
- Department of Biotechnology, Maebashi Institute of Technology, 460-1 Kamisadori-machi, Maebashi, Gunma, 371-0816, Japan
| | - Koyo Koda
- Department of Biotechnology, Maebashi Institute of Technology, 460-1 Kamisadori-machi, Maebashi, Gunma, 371-0816, Japan
| | - Kin-Ya Kubo
- Seijoh University Graduate School of Health Care Studies, 2-172 Fukinodai, Tokai, Aichi, 476-8588, Japan
| | - Toshiharu Yamamoto
- Brain Functions and Neuroscience Division, Department of Oral Science, Kanagawa Dental University Graduate School, Inaoka-cho 82, Yokosuka, Kanagawa, 238-0003, Japan
| | - Toshihiko Kadoya
- Department of Biotechnology, Maebashi Institute of Technology, 460-1 Kamisadori-machi, Maebashi, Gunma, 371-0816, Japan
| |
Collapse
|
28
|
Zeng Q, Liu S, Yao J, Zhang Y, Yuan Z, Jiang C, Chen A, Fu Q, Su B, Dunham R, Liu Z. Transcriptome Display During Testicular Differentiation of Channel Catfish (Ictalurus punctatus) as Revealed by RNA-Seq Analysis. Biol Reprod 2016; 95:19. [PMID: 27307075 DOI: 10.1095/biolreprod.116.138818] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/26/2016] [Indexed: 12/13/2022] Open
Abstract
Channel catfish (Ictalurus punctatus) has been recognized as a dominant freshwater aquaculture species in the United States. It is also a suitable model for studying the mechanisms of sex determination and differentiation because of its sexual plasticity and exhibition of both genetic and environmental sex determination. The testicular differentiation in male channel catfish normally starts between 90 and 102 days postfertilization (dpf), while the ovarian differentiation starts early from 19 dpf. As such, efforts to better understand the postponed testicular development at the molecular level are needed. Toward that end, we conducted transcriptomic comparison of gene expression of male and female gonads at 90, 100, and 110 dpf using high-throughput RNA-Seq. Transcriptomic profiles of male gonads on 90 and 100 dpf exhibited high similarities except for a small number of significantly up-regulated genes that were involved in development of germ cell-supporting somatic cells, while drastic changes were observed during 100-110 dpf, with a group of highly up-regulated genes that were involved in germ cells development, including nanog and pou5f1 Transcriptomic comparison between testes and ovaries identified male-preferential genes, such as gsdf, cxcl12, as well as other cytokines mediated the development of the gonad into a testis. Co-expression analysis revealed highly correlated genes and potential pathways underlying germ cell differentiation and spermatogonia stem cell development. The candidate genes and pathways identified in this study set the foundation for further studies on sex determination and differentiation in catfish as well as other teleosts.
Collapse
Affiliation(s)
- Qifan Zeng
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, and Program of Cell and Molecular Biosciences, Auburn, Alabama
| | - Shikai Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, and Program of Cell and Molecular Biosciences, Auburn, Alabama
| | - Jun Yao
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, and Program of Cell and Molecular Biosciences, Auburn, Alabama
| | - Yu Zhang
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, and Program of Cell and Molecular Biosciences, Auburn, Alabama
| | - Zihao Yuan
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, and Program of Cell and Molecular Biosciences, Auburn, Alabama
| | - Chen Jiang
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, and Program of Cell and Molecular Biosciences, Auburn, Alabama
| | - Ailu Chen
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, and Program of Cell and Molecular Biosciences, Auburn, Alabama
| | - Qiang Fu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, and Program of Cell and Molecular Biosciences, Auburn, Alabama
| | - Baofeng Su
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, and Program of Cell and Molecular Biosciences, Auburn, Alabama
| | - Rex Dunham
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, and Program of Cell and Molecular Biosciences, Auburn, Alabama
| | - Zhanjiang Liu
- The Fish Molecular Genetics and Biotechnology Laboratory, Aquatic Genomics Unit, School of Fisheries, Aquaculture and Aquatic Sciences, and Program of Cell and Molecular Biosciences, Auburn, Alabama
| |
Collapse
|
29
|
Thiemann S, Baum LG. Galectins and Immune Responses—Just How Do They Do Those Things They Do? Annu Rev Immunol 2016; 34:243-64. [DOI: 10.1146/annurev-immunol-041015-055402] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sandra Thiemann
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California 90095; ,
| | - Linda G. Baum
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California 90095; ,
| |
Collapse
|
30
|
Restuccia A, Fettis MM, Hudalla GA. Glycomaterials for immunomodulation, immunotherapy, and infection prophylaxis. J Mater Chem B 2016; 4:1569-1585. [DOI: 10.1039/c5tb01780g] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Synthetic carbohydrate-modified materials that can engage the innate and adaptive immune systems are receiving increasing interest to confer protection against onset of future disease, such as pathogen infection, as well as to treat established diseases, such as autoimmunity and cancer.
Collapse
Affiliation(s)
- Antonietta Restuccia
- J. Crayton Pruitt Family Department of Biomedical Engineering
- University of Florida
- Gainesville
- USA
| | - Margaret M. Fettis
- J. Crayton Pruitt Family Department of Biomedical Engineering
- University of Florida
- Gainesville
- USA
| | - Gregory A. Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering
- University of Florida
- Gainesville
- USA
| |
Collapse
|
31
|
Thiemann S, Man JH, Chang MH, Lee B, Baum LG. Galectin-1 regulates tissue exit of specific dendritic cell populations. J Biol Chem 2015. [PMID: 26216879 PMCID: PMC4566239 DOI: 10.1074/jbc.m115.644799] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
During inflammation, dendritic cells emigrate from inflamed tissue across the lymphatic endothelium into the lymphatic vasculature and travel to regional lymph nodes to initiate immune responses. However, the processes that regulate dendritic cell tissue egress and migration across the lymphatic endothelium are not well defined. The mammalian lectin galectin-1 is highly expressed by vascular endothelial cells in inflamed tissue and has been shown to regulate immune cell tissue entry into inflamed tissue. Here, we show that galectin-1 is also highly expressed by human lymphatic endothelial cells, and deposition of galectin-1 in extracellular matrix selectively regulates migration of specific human dendritic cell subsets. The presence of galectin-1 inhibits migration of immunogenic dendritic cells through the extracellular matrix and across lymphatic endothelial cells, but it has no effect on migration of tolerogenic dendritic cells. The major galectin-1 counter-receptor on both dendritic cell populations is the cell surface mucin CD43; differential core 2 O-glycosylation of CD43 between immunogenic dendritic cells and tolerogenic dendritic cells appears to contribute to the differential effect of galectin-1 on migration. Binding of galectin-1 to immunogenic dendritic cells reduces phosphorylation and activity of the protein-tyrosine kinase Pyk2, an effect that may also contribute to reduced migration of this subset. In a murine lymphedema model, galectin-1(-/-) animals had increased numbers of migratory dendritic cells in draining lymph nodes, specifically dendritic cells with an immunogenic phenotype. These findings define a novel role for galectin-1 in inhibiting tissue emigration of immunogenic, but not tolerogenic, dendritic cells, providing an additional mechanism by which galectin-1 can dampen immune responses.
Collapse
Affiliation(s)
- Sandra Thiemann
- From the Departments of Pathology and Laboratory Medicine and
| | - Jeanette H Man
- From the Departments of Pathology and Laboratory Medicine and
| | - Margaret H Chang
- Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California 90095 and
| | - Benhur Lee
- From the Departments of Pathology and Laboratory Medicine and Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California 90095 and the Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Linda G Baum
- From the Departments of Pathology and Laboratory Medicine and
| |
Collapse
|
32
|
Norozi F, Ahmadzadeh A, Shahjahani M, Shahrabi S, Saki N. Twist as a new prognostic marker in hematological malignancies. Clin Transl Oncol 2015. [DOI: 10.1007/s12094-015-1357-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
33
|
Assembly, organization and regulation of cell-surface receptors by lectin–glycan complexes. Biochem J 2015; 469:1-16. [DOI: 10.1042/bj20150461] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Galectins are a family of β-galactoside-binding lectins carrying at least one consensus sequence in the carbohydrate-recognition domain. Properties of glycosylated ligands, such as N- and O-glycan branching, LacNAc (N-acetyl-lactosamine) content and the balance of α2,3- and α2,6-linked sialic acid dramatically influence galectin binding to a preferential set of counter-receptors. The presentation of specific glycans in galectin-binding partners is also critical, as proper orientation and clustering of oligosaccharide ligands on multiple carbohydrate side chains increase the binding avidity of galectins for particular glycosylated receptors. When galectins are released from the cells, they typically concentrate on the cell surface and the local matrix, raising their local concentration. Thus galectins can form their own multimers in the extracellular milieu, which in turn cross-link glycoconjugates on the cell surface generating galectin–glycan complexes that modulate intracellular signalling pathways, thus regulating cellular processes such as apoptosis, proliferation, migration and angiogenesis. Subtle changes in receptor expression, rates of protein synthesis, activities of Golgi enzymes, metabolite concentrations supporting glycan biosynthesis, density of glycans, strength of protein–protein interactions at the plasma membrane and stoichiometry may modify galectin–glycan complexes. Although galectins are key contributors to the formation of these extended glycan complexes leading to promotion of receptor segregation/clustering, and inhibition of receptor internalization by surface retention, when these complexes are disrupted, some galectins, particularly galectin-3 and -4, showed the ability to drive clathrin-independent mechanisms of endocytosis. In the present review, we summarize the data available on the assembly, hierarchical organization and regulation of conspicuous galectin–glycan complexes, and their implications in health and disease.
Collapse
|
34
|
Induction of Apoptosis and Antitumor Activity of Eel Skin Mucus, Containing Lactose-Binding Molecules, on Human Leukemic K562 Cells. Mar Drugs 2015; 13:3936-49. [PMID: 26090845 PMCID: PMC4483664 DOI: 10.3390/md13063936] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 06/02/2015] [Accepted: 06/05/2015] [Indexed: 12/23/2022] Open
Abstract
For innate immune defense, lower animals such as fish and amphibian are covered with skin mucus, which acts as both a mechanical and biochemical barrier. Although several mucus sources have been isolated and studied for their biochemical and immunological functions, the precise mechanism(s) of action remains unknown. In the present study, we additionally found the eel skin mucus (ESM) to be a promising candidate for use in anti-tumor therapy. Our results showed that the viability of K562 cells was decreased in a dose-dependent manner by treatment with the isolated ESM. The cleaved forms of caspase-9, caspase-3 and poly adenosine diphosphate-ribose polymerase were increased by ESM. The levels of Bax expression and released cytochrome C were also increased after treatment with ESM. Furthermore, during the ESM mediated-apoptosis, phosphorylation levels of ERK1/2 and p38 but not JNK were increased and cell viabilities of the co-treated cells with ESM and inhibitors of ERK 1/2 or p38 were also increased. In addition, treatment with lactose rescued the ESM-mediated decrease in cell viability, indicating lactose-containing glycans in the leukemia cells acted as a counterpart of the ESM for interaction. Taken together, these results suggest that ESM could induce mitochondria-mediated apoptosis through membrane interaction of the K562 human leukemia cells. To the best of our knowledge, this is the first observation that ESM has anti-tumor activity in human cells.
Collapse
|
35
|
Garner OB, Yun T, Pernet O, Aguilar HC, Park A, Bowden TA, Freiberg AN, Lee B, Baum LG. Timing of galectin-1 exposure differentially modulates Nipah virus entry and syncytium formation in endothelial cells. J Virol 2015; 89:2520-9. [PMID: 25505064 PMCID: PMC4325760 DOI: 10.1128/jvi.02435-14] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/03/2014] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED Nipah virus (NiV) is a deadly emerging enveloped paramyxovirus that primarily targets human endothelial cells. Endothelial cells express the innate immune effector galectin-1 that we have previously shown can bind to specific N-glycans on the NiV envelope fusion glycoprotein (F). NiV-F mediates fusion of infected endothelial cells into syncytia, resulting in endothelial disruption and hemorrhage. Galectin-1 is an endogenous carbohydrate-binding protein that binds to specific glycans on NiV-F to reduce endothelial cell fusion, an effect that may reduce pathophysiologic sequelae of NiV infection. However, galectins play multiple roles in regulating host-pathogen interactions; for example, galectins can promote attachment of HIV to T cells and macrophages and attachment of HSV-1 to keratinocytes but can also inhibit influenza entry into airway epithelial cells. Using live Nipah virus, in the present study, we demonstrate that galectin-1 can enhance NiV attachment to and infection of primary human endothelial cells by bridging glycans on the viral envelope to host cell glycoproteins. In order to exhibit an enhancing effect, galectin-1 must be present during the initial phase of virus attachment; in contrast, addition of galectin-1 postinfection results in reduced production of progeny virus and syncytium formation. Thus, galectin-1 can have dual and opposing effects on NiV infection of human endothelial cells. While various roles for galectin family members in microbial-host interactions have been described, we report opposing effects of the same galectin family member on a specific virus, with the timing of exposure during the viral life cycle determining the outcome. IMPORTANCE Nipah virus is an emerging pathogen that targets endothelial cells lining blood vessels; the high mortality rate (up to 70%) in Nipah virus infections results from destruction of these cells and resulting catastrophic hemorrhage. Host factors that promote or prevent Nipah virus infection are not well understood. Endogenous human lectins, such as galectin-1, can function as pattern recognition receptors to reduce infection and initiate immune responses; however, lectins can also be exploited by microbes to enhance infection of host cells. We found that galectin-1, which is made by inflamed endothelial cells, can both promote Nipah virus infection of endothelial cells by "bridging" the virus to the cell, as well as reduce production of progeny virus and reduce endothelial cell fusion and damage, depending on timing of galectin-1 exposure. This is the first report of spatiotemporal opposing effects of a host lectin for a virus in one type of host cell.
Collapse
Affiliation(s)
- Omai B Garner
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Tatyana Yun
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Olivier Pernet
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Hector C Aguilar
- Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Arnold Park
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Thomas A Bowden
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Alexander N Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Benhur Lee
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California, USA Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Linda G Baum
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
36
|
Pena C, Mirandola L, Figueroa JA, Hosiriluck N, Suvorava N, Trotter K, Reidy A, Rakhshanda R, Payne D, Jenkins M, Grizzi F, Littlefield L, Chiriva-Internati M, Cobos E. Galectins as therapeutic targets for hematological malignancies: a hopeful sweetness. ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:87. [PMID: 25405162 DOI: 10.3978/j.issn.2305-5839.2014.09.14] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 09/20/2014] [Indexed: 01/04/2023]
Abstract
Galectins are family of galactose-binding proteins known to play critical roles in inflammation and neoplastic progression. Galectins facilitate the growth and survival of neoplastic cells by regulating their cross-talk with the extracellular microenvironment and hampering anti-neoplastic immunity. Here, we review the role of galectins in the biology of hematological malignancies and their promise as potential therapeutic agents in these diseases.
Collapse
Affiliation(s)
- Camilo Pena
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Leonardo Mirandola
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Jose A Figueroa
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Nattamol Hosiriluck
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Natallia Suvorava
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Kayley Trotter
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Adair Reidy
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Rahman Rakhshanda
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Drew Payne
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Marjorie Jenkins
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Fabio Grizzi
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Lauren Littlefield
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Maurizio Chiriva-Internati
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| | - Everardo Cobos
- 1 Department of Internal Medicine at the Division of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX, USA ; 2 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 3 Kiromic LLC, TX, USA ; 4 Division of Surgical Oncology, Texas Tech University Medical Center, Amarillo, TX, USA ; 5 Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Amarillo, TX, USA ; 6 Humanitas Clinical and Research Center, Milan, Italy
| |
Collapse
|
37
|
Ruggeri RR, Bressan FF, Siqueira NM, Meirelles F, Frantz N, Watanabe YF, Soares RMD, Bos-Mikich A. Derivation and culture of putative parthenogenetic embryonic stem cells in new gelatin substrates modified with galactomannan. Macromol Res 2014. [DOI: 10.1007/s13233-014-2151-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
38
|
Cheng DE, Chang WA, Hung JY, Huang MS, Kuo PL. Involvement of IL‑10 and granulocyte colony‑stimulating factor in the fate of monocytes controlled by galectin‑1. Mol Med Rep 2014; 10:2389-94. [PMID: 25231117 DOI: 10.3892/mmr.2014.2573] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 06/05/2014] [Indexed: 11/05/2022] Open
Abstract
The process of differentiation from monocytes to dendritic cells is critical in immune modulation. Monocyte apoptosis is a key regulator in balancing the immune response. Galectin‑1 has been reported to induce tolerogenic dendritic cells by the autocrine interleukin (IL)‑10 in monocytes. However, IL‑10 has been found to induce apoptosis in IL‑4/granulocyte macrophage colony‑stimulating factor (CSF) stimulating and non‑stimulating monocytes, whereas galectin‑1 has not. After analyzing the factors secreted by galectin-1-activated CD14 monocytes isolated from the peripheral blood, the present study revealed that galectin‑1 upregulates IL‑10 and granulocyte (G)-CSF expression. Furthermore, G‑CSF inhibited IL‑10‑induced apoptosis, implying that galectin‑1 may enhance the immune‑modulating functions of G‑CSF by inducing tolerogenic dendritic cells and maintaining their survival. Therefore, G‑CSF may be further applied in immune therapy, particularly in the IL‑10‑presenting microenvironment.
Collapse
Affiliation(s)
- Da-En Cheng
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Wei-An Chang
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Jen-Yu Hung
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Ming-Shyan Huang
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Po-Lin Kuo
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
39
|
Abstract
Radiation therapy is a main stay in treating solid tumors and plays a significant role in definitive and adjuvant therapy. Unfortunately, local control remains a challenge, in which the success of radiotherapy is largely dictated by tumor hypoxia, DNA damage repair and the antitumor immune response. Extensive efforts have therefore been devoted to targeting the factors that attenuate tumor radiosensitivity, although with limited success. Mounting evidence suggests that tumor and endothelial cells may utilize galectin-1 (Gal-1) for protection against radiation through several mechanisms. Targeting Gal-1 in combination with radiotherapy provides an exciting approach to address several radiation-prohibitive mechanisms.
Collapse
Affiliation(s)
- Peiwen Kuo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
40
|
Rachel H, Chang-Chun L. Recent advances toward the development of inhibitors to attenuate tumor metastasis via the interruption of lectin-ligand interactions. Adv Carbohydr Chem Biochem 2014; 69:125-207. [PMID: 24274369 DOI: 10.1016/b978-0-12-408093-5.00005-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aberrant glycosylation is a well-recognized phenomenon that occurs on the surface of tumor cells, and the overexpression of a number of ligands (such as TF, sialyl Tn, and sialyl Lewis X) has been correlated to a worse prognosis for the patient. These unique carbohydrate structures play an integral role in cell-cell communication and have also been associated with more metastatic cancer phenotypes, which can result from binding to lectins present on cell surfaces. The most well studied metastasis-associated lectins are the galectins and selectins, which have been correlated to adhesion, neoangiogenesis, and immune-cell evasion processes. In order to slow the rate of metastatic lesion formation, a number of approaches have been successfully developed which involve interfering with the tumor lectin-substrate binding event. Through the generation of inhibitors, or by attenuating lectin and/or carbohydrate expression, promising results have been observed both in vitro and in vivo. This article briefly summarizes the involvement of lectins in the metastatic process and also describes different approaches used to prevent these undesirable carbohydrate-lectin binding events, which should ultimately lead to improvement in current cancer therapies.
Collapse
Affiliation(s)
- Hevey Rachel
- Alberta Glycomics Centre, Department of Chemistry, University of Calgary, Calgary, Alberta, Canada
| | | |
Collapse
|
41
|
Vladoiu MC, Labrie M, St-Pierre Y. Intracellular galectins in cancer cells: potential new targets for therapy (Review). Int J Oncol 2014; 44:1001-14. [PMID: 24452506 DOI: 10.3892/ijo.2014.2267] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 12/02/2013] [Indexed: 11/06/2022] Open
Abstract
Dysregulation of galectin expression is frequently observed in cancer tissues. Such an abnormal expression pattern often correlates with aggressiveness and relapse in many types of cancer. Because galectins have the ability to modulate functions that are important for cell survival, migration and metastasis, they also represent attractive targets for cancer therapy. This has been well-exploited for extracellular galectins, which bind glycoconjugates expressed on the surface of cancer cells. Although the existence of intracellular functions of galectins has been known for many years, an increasing number of studies indicate that these proteins can also alter tumor progression through their interaction with intracellular ligands. In fact, in some instances, the interactions of galectins with their intracellular ligands seem to occur independently of their carbohydrate recognition domain. Such findings call for a change in the basic assumptions, or paradigms, concerning the activity of galectins in cancer and may force us to revisit our strategies to develop galectin antagonists for the treatment of cancer.
Collapse
Affiliation(s)
| | | | - Yves St-Pierre
- INRS-Institut Armand-Frappier, Laval, QC H7V 1B7, Canada
| |
Collapse
|
42
|
Anginot A, Espeli M, Chasson L, Mancini SJC, Schiff C. Galectin 1 modulates plasma cell homeostasis and regulates the humoral immune response. THE JOURNAL OF IMMUNOLOGY 2013; 190:5526-33. [PMID: 23616571 DOI: 10.4049/jimmunol.1201885] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Galectin-1 (GAL1) is an S-type lectin with multiple functions, including the control of B cell homeostasis. GAL1 expression was reported to be under the control of the plasma cell master regulator BLIMP-1. GAL1 was detected at the protein level in LPS-stimulated B cells and was shown to promote Ig secretion in vitro. However, the pattern of GAL1 expression and function of GAL1 in B cells in vivo are still unclear. In this study, we show that, among B cells, GAL1 is only expressed by differentiating plasma cells following T-dependent or T-independent immunization. Using GAL1-deficient mice we demonstrate that GAL1 expression is required for the maintenance of Ag-specific Ig titers and Ab-secreting cell numbers. Using an in vitro differentiation assay we find that GAL1-deficient plasmablasts can develop normally but die rapidly, through caspase 8 activation, under serum starvation-induced death conditions. TUNEL assays show that in vivo-generated GAL1-deficient plasma cells exhibit an increased sensitivity to apoptosis. Taken together, our data indicate that endogenous GAL1 supports plasma cell survival and participates in the regulation of the humoral immune response.
Collapse
Affiliation(s)
- Adrienne Anginot
- Centre d'Immunologie de Marseille-Luminy, Faculté des Sciences de Luminy, Aix Marseille University, UM2, Marseille F-13288, France.
| | | | | | | | | |
Collapse
|
43
|
Abrudan J, Ramalho-Ortigão M, O'Neil S, Stayback G, Wadsworth M, Bernard M, Shoue D, Emrich S, Lawyer P, Kamhawi S, Rowton ED, Lehane MJ, Bates PA, Valenzeula JG, Tomlinson C, Appelbaum E, Moeller D, Thiesing B, Dillon R, Clifton S, Lobo NF, Wilson RK, Collins FH, McDowell MA. The characterization of the Phlebotomus papatasi transcriptome. INSECT MOLECULAR BIOLOGY 2013; 22:211-232. [PMID: 23398403 PMCID: PMC3594503 DOI: 10.1111/imb.12015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
As important vectors of human disease, phlebotomine sand flies are of global significance to human health, transmitting several emerging and re-emerging infectious diseases. The most devastating of the sand fly transmitted infections are the leishmaniases, causing significant mortality and morbidity in both the Old and New World. Here we present the first global transcriptome analysis of the Old World vector of cutaneous leishmaniasis, Phlebotomus papatasi (Scopoli) and compare this transcriptome to that of the New World vector of visceral leishmaniasis, Lutzomyia longipalpis. A normalized cDNA library was constructed using pooled mRNA from Phlebotomus papatasi larvae, pupae, adult males and females fed sugar, blood, or blood infected with Leishmania major. A total of 47 615 generated sequences was cleaned and assembled into 17 120 unique transcripts. Of the assembled sequences, 50% (8837 sequences) were classified using Gene Ontology (GO) terms. This collection of transcripts is comprehensive, as demonstrated by the high number of different GO categories. An in-depth analysis revealed 245 sequences with putative homology to proteins involved in blood and sugar digestion, immune response and peritrophic matrix formation. Twelve of the novel genes, including one trypsin, two peptidoglycan recognition proteins (PGRP) and nine chymotrypsins, have a higher expression level during larval stages. Two novel chymotrypsins and one novel PGRP are abundantly expressed upon blood feeding. This study will greatly improve the available genomic resources for P. papatasi and will provide essential information for annotation of the full genome.
Collapse
Affiliation(s)
- Jenica Abrudan
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Marcelo Ramalho-Ortigão
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | | | | | | | | | | | | | - Phillip Lawyer
- Intracellular Parasite Biology Section, Laboratory of Parasitic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Edgar D. Rowton
- Entomology Program, Walter Reed Army Institute of Research, 530 Robert Grant Ave., Silver Spring, MD 20910, USA
| | | | - Paul A. Bates
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, LA1 4YQ, UK
| | - Jesus G. Valenzeula
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Chad Tomlinson
- The Genome Institute at Washington University, St. Louis, Missouri, 63108, USA
| | - Elizabeth Appelbaum
- The Genome Institute at Washington University, St. Louis, Missouri, 63108, USA
| | - Deborah Moeller
- The Genome Institute at Washington University, St. Louis, Missouri, 63108, USA
| | - Brenda Thiesing
- The Genome Institute at Washington University, St. Louis, Missouri, 63108, USA
| | - Rod Dillon
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, LA1 4YQ, UK
| | - Sandra Clifton
- The Genome Institute at Washington University, St. Louis, Missouri, 63108, USA
| | - Neil F. Lobo
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Richard K. Wilson
- The Genome Institute at Washington University, St. Louis, Missouri, 63108, USA
| | - Frank H. Collins
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Mary Ann McDowell
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
44
|
Abstract
Rheumatoid arthritis (RA) is a complex and common systemic autoimmune disease characterized by synovial inflammation and hyperplasia. Multiple proteins, cells, and pathways have been identified to contribute to the pathogenesis of RA. Galectins are a group of lectins that bind to β-galactoside carbohydrates on the cell surface and in the extracellular matrix. They are expressed in a wide variety of tissues and organs with the highest expression in the immune system. Galectins are potent immune regulators and modulate a range of pathological processes, such as inflammation, autoimmunity, and cancer. Accumulated evidence shows that several family members of galectins play positive or negative roles in the disease development of RA, through their effects on T and B lymphocytes, myeloid lineage cells, and fibroblast-like synoviocytes. In this review, we will summarize the function of different galectins in immune modulation and their distinct roles in RA pathogenesis.
Collapse
Affiliation(s)
- Song Li
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yangsheng Yu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Christopher D Koehn
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zhixin Zhang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA ; The Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kaihong Su
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA ; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA ; The Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
45
|
Jouve N, Despoix N, Espeli M, Gauthier L, Cypowyj S, Fallague K, Schiff C, Dignat-George F, Vély F, Leroyer AS. The involvement of CD146 and its novel ligand Galectin-1 in apoptotic regulation of endothelial cells. J Biol Chem 2012; 288:2571-9. [PMID: 23223580 DOI: 10.1074/jbc.m112.418848] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
CD146 is a highly glycosylated junctional adhesion molecule, expressed on human vascular endothelial cells and involved in the control of vessel integrity. Galectin-1 is a lectin produced by vascular cells that can binds N- and O-linked oligosaccharides of cell membrane glycoproteins. Because both CD146 and Galectin-1 are involved in modulation of cell apoptosis, we hypothesized that Galectin-1 could interact with CD146, leading to functional consequences in endothelial cell apoptosis. We first characterized CD146 glycosylations and showed that it is mainly composed of N-glycans able to establish interactions with Galectin-1. We demonstrated a sugar-dependent binding of recombinant CD146 to Galectin-1 using both ELISA and Biacore assays. This interaction is direct, with a K(D) of 3.10(-7) M, and specific as CD146 binds to Galectin-1 and not to Galectin-2. Moreover, co-immunoprecipitation experiments showed that Galectin-1 interacts with endogenous CD146 that is highly expressed by HUVEC. We observed a Galectin-1-induced HUVEC apoptosis in a dose-dependent manner as demonstrated by Annexin-V/7AAD staining. Interestingly, both down-regulation of CD146 cell surface expression using siRNA and antibody-mediated blockade of CD146 increase this apoptosis. Altogether, our results identify Galectin-1 as a novel ligand for CD146 and this interaction protects, in vitro, endothelial cells against apoptosis induced by Galectin-1.
Collapse
Affiliation(s)
- Nathalie Jouve
- Aix-Marseille Université, INSERM, UMR-S 1076, 13385 Marseille, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Auvynet C, Moreno S, Melchy E, Coronado-Martínez I, Montiel JL, Aguilar-Delfin I, Rosenstein Y. Galectin-1 promotes human neutrophil migration. Glycobiology 2012; 23:32-42. [PMID: 22942212 DOI: 10.1093/glycob/cws128] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
An important step of innate immune response is the recruitment of polymorphonuclear leukocytes (PMN) to injured tissues through chemotactic molecules. Galectins, a family of endogenous lectins, participate in numerous functions such as lymphoid cell migration, homing, cell-cell and cell-matrix interactions. Particularly, galectin-3 (Gal-3) and -9 have been implicated in the modulation of acute and chronic inflammation by inducing the directional migration of monocytes/macrophages and eosinophils, whereas Gal-1 is considered to function as an anti-inflammatory molecule, capable of inhibiting the influx of PMN to the site of injury. In this study, we assessed the effect of Gal-1 on neutrophil recruitment, in the absence of additional inflammatory insults. Contrasting with its capacity to inhibit cell trafficking and modulate the release of mediators described in models of acute inflammation and autoimmunity, we evidenced that Gal-1 has the capacity to induce neutrophil migration both in vitro and in vivo. This effect is not mediated through a G-protein-coupled receptor but potentially through the sialoglycoprotein CD43, via carbohydrate binding and through the p38 mitogen-activated protein kinase pathway. These results suggest a novel biological function for CD43 on neutrophils and highlight that depending on the environment, Gal-1 can act either as chemoattractant or, as a molecule that negatively regulates migration under acute inflammatory conditions, underscoring the potential of Gal-1 as a target for innovative drug development.
Collapse
Affiliation(s)
- Constance Auvynet
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnologia, Universidad Nacional Autónoma de México, Avenida Universidad 2001, Col Chamilpa, Cuernavaca, Morelos 62210, Mexico
| | | | | | | | | | | | | |
Collapse
|
47
|
Miller MC, Klyosov AA, Mayo KH. Structural features for α-galactomannan binding to galectin-1. Glycobiology 2012; 22:543-51. [PMID: 22156919 PMCID: PMC3287016 DOI: 10.1093/glycob/cwr173] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 11/04/2011] [Accepted: 11/20/2011] [Indexed: 12/16/2022] Open
Abstract
Galectins have a highly conserved carbohydrate-binding domain to which a variety of galactose-containing saccharides, both β- and α-galactosides, can interact with varying degrees of affinity. Recently, we demonstrated that the relatively large α(1 → 6)-D-galacto-β(1 → 4)-D-mannan (Davanat) binds galectin-1 (gal-1) primarily at an alternative carbohydrate-binding domain. Here, we used a series of α-galactomannans (GMs) that vary in their mannose-to-galactose ratios for insight into an optimal structural signature for GM binding to gal-1. Heteronuclear single-quantum coherence nuclear magnetic resonance spectroscopy with (15)N-labeled gal-1 and statistical modeling suggest that the optimal signature consists of α-D-galactopyranosyl doublets surrounded by regions of about four or more "naked" mannose residues. These relatively large and complex GMs all appear to interact with varying degrees at essentially the same binding surface on gal-1 that includes the Davanat alternative binding site and elements of the canonical β-galactoside-binding region. The use of two small, well-defined GMs [6(1)-α(1 → 6)-D-galactosyl-β-D-mannotriaose and 6(3),6(4)-di-α(1 → 6)-D-galactosyl-β-D-mannopentaose] helped characterize how GMs, in general, interact in part with the canonical site. Overall, our findings contribute to better understanding interactions of gal-1 with larger, complex polysaccharides and to the development of GM-based therapeutics for clinical use.
Collapse
Affiliation(s)
- Michelle C Miller
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Health Sciences Center, 6-155 Jackson Hall, 321 Church Street, Minneapolis, MN 55455, USA
| | | | - Kevin H Mayo
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Health Sciences Center, 6-155 Jackson Hall, 321 Church Street, Minneapolis, MN 55455, USA
| |
Collapse
|
48
|
Tribulatti MV, Figini MG, Carabelli J, Cattaneo V, Campetella O. Redundant and Antagonistic Functions of Galectin-1, -3, and -8 in the Elicitation of T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2012; 188:2991-9. [DOI: 10.4049/jimmunol.1102182] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
49
|
Clark MC, Baum LG. T cells modulate glycans on CD43 and CD45 during development and activation, signal regulation, and survival. Ann N Y Acad Sci 2012; 1253:58-67. [PMID: 22288421 DOI: 10.1111/j.1749-6632.2011.06304.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Glycosylation affects many essential T cell processes and is intrinsically controlled throughout the lifetime of a T cell. CD43 and CD45 are the two most abundant glycoproteins on the T cell surface and are decorated with O- and N-glycans. Global T cell glycosylation and specific glycosylation of CD43 and CD45 are modulated during thymocyte development and T cell activation; T cells control the type and abundance of glycans decorating CD43 and CD45 by regulating expression of glycosyltransferases and glycosidases. Additionally, T cells regulate glycosylation of CD45 by expressing alternatively spliced isoforms of CD45 that have different glycan attachment sites. The glycophenotype of CD43 and CD45 on T cells influences how T cells interact with the extracellular environment, including how T cells interact with endogenous lectins. This review focuses on changes in glycosylation of CD43 and CD45 occurring throughout T cell development and activation and the role that glycosylation plays in regulating T cell processes, such as migration, T cell receptor signaling, and apoptosis.
Collapse
Affiliation(s)
- Mary C Clark
- Department of Pathology and Laboratory Medicine, UCLA School of Medicine, University of California, Los Angeles, USA
| | | |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Galectins, a family of evolutionarily conserved glycan-binding proteins, are involved in the regulation of multiple cellular processes (e.g. immunity, apoptosis, cellular signaling, development, angiogenesis and cellular growth) and diseases (e.g. chronic inflammation, autoimmunity, cancer, infection). We discuss here how galectins contribute to the development of specialized microenvironmental niches during hematopoiesis. RECENT FINDINGS An expanding set of data strengthens a role of galectins in hematopoietic differentiation, particularly by setting specific interactions between hematopoietic and stromal cells: galectin-5 is found in reticulocytes and erythroblastic islands suggesting a major role during erythropoiesis; galectin-1 and 3 are involved in thymocyte apoptosis, signaling and intrathymic migration; galectin-1 plays critical roles in pre-BII cells development. Moreover, expression of galectins-1 and 10 are differentially expressed during T-regulatory cell development. Various galectins (3, 4, 5, 9) have been reported to be regulated during myelopoiesis and traffic into intracellular compartments, dictating the cellular distribution of specific glycoproteins and glycosphingolipids. SUMMARY The abundance of galectins in both extracellular and intracellular compartments, their multifunctional properties and ability to form supramolecular signaling complexes with specific glycoconjugates, make these glycan-binding proteins excellent candidates to mediate interactions between hematopoietic cells and the stromal microenvironment. Their secretion by one of the cellular partners can modulate adhesive properties by cross-linking specific glycoconjugates present on stromal or hematopoietic cells, by favoring the formation of synapses or by creating glycoprotein lattices on the surface of different cell types. Their divergent specificities and affinities for various glycoproteins contribute to the multiplicity of their cellular interactions.
Collapse
|