1
|
Zhang N, Liu Q, Wang D, Wang X, Pan Z, Han B, He G. Multifaceted roles of Galectins: from carbohydrate binding to targeted cancer therapy. Biomark Res 2025; 13:49. [PMID: 40134029 PMCID: PMC11934519 DOI: 10.1186/s40364-025-00759-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025] Open
Abstract
Galectins play pivotal roles in cellular recognition and signaling processes by interacting with glycoconjugates. Extensive research has highlighted the significance of Galectins in the context of cancer, aiding in the identification of biomarkers for early detection, personalized therapy, and predicting treatment responses. This review offers a comprehensive overview of the structural characteristics, ligand-binding properties, and interacting proteins of Galectins. We delve into their biological functions and examine their roles across various cancer types. Galectins, characterized by a conserved carbohydrate recognition domain (CRD), are divided into prototype, tandem-repeat, and chimera types based on their structural configurations. Prototype Galectins contain a single CRD, tandem-repeat Galectins contain two distinct CRDs linked by a peptide, and the chimera-type Galectin-3 features a unique structural arrangement. The capacity of Galectins to engage in multivalent interactions allows them to regulate a variety of signaling pathways, thereby affecting cell fate and function. In cancer, Galectins contribute to tumor cell transformation, angiogenesis, immune evasion, and metastasis, making them critical targets for therapeutic intervention. This review discusses the multifaceted roles of Galectins in cancer progression and explores current advancements in the development of Galectin-targeted therapies. We also address the challenges and future directions for integrating Galectin research into clinical practice to enhance cancer treatment outcomes. In brief, understanding the complex functions of Galectins in cancer biology opens new avenues for therapeutic strategies. Continued research on Galectin interactions and their pathological roles is essential for developing effective carbohydrate-based treatments and improving clinical interventions for cancer patients.
Collapse
Affiliation(s)
- Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Qiao Liu
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Daihan Wang
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Xiaoyun Wang
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Zhaoping Pan
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Medical Technology and School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Gu He
- Institute of Precision Drug Innovation and Cancer Center, the Second Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
2
|
Perez-Moreno E, Oyanadel C, de la Peña A, Hernández R, Pérez-Molina F, Metz C, González A, Soza A. Galectins in epithelial-mesenchymal transition: roles and mechanisms contributing to tissue repair, fibrosis and cancer metastasis. Biol Res 2024; 57:14. [PMID: 38570874 PMCID: PMC10993482 DOI: 10.1186/s40659-024-00490-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/12/2024] [Indexed: 04/05/2024] Open
Abstract
Galectins are soluble glycan-binding proteins that interact with a wide range of glycoproteins and glycolipids and modulate a broad spectrum of physiological and pathological processes. The expression and subcellular localization of different galectins vary among tissues and cell types and change during processes of tissue repair, fibrosis and cancer where epithelial cells loss differentiation while acquiring migratory mesenchymal phenotypes. The epithelial-mesenchymal transition (EMT) that occurs in the context of these processes can include modifications of glycosylation patterns of glycolipids and glycoproteins affecting their interactions with galectins. Moreover, overexpression of certain galectins has been involved in the development and different outcomes of EMT. This review focuses on the roles and mechanisms of Galectin-1 (Gal-1), Gal-3, Gal-4, Gal-7 and Gal-8, which have been involved in physiologic and pathogenic EMT contexts.
Collapse
Affiliation(s)
- Elisa Perez-Moreno
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia (CCTE) Ciencia y Vida, Santiago, Chile
| | - Claudia Oyanadel
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Adely de la Peña
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia (CCTE) Ciencia y Vida, Santiago, Chile
| | - Ronny Hernández
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Francisca Pérez-Molina
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Claudia Metz
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Alfonso González
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.
- Centro Científico y Tecnológico de Excelencia (CCTE) Ciencia y Vida, Santiago, Chile.
| | - Andrea Soza
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.
- Centro Científico y Tecnológico de Excelencia (CCTE) Ciencia y Vida, Santiago, Chile.
| |
Collapse
|
3
|
Pinto NA, Abba MC, Laporte L, Pérez Sáez JM, Blidner AG, Torres NI, Morales RM, Gatto SG, Bach CA, Veigas F, García Rivello HJ, Song P, Frederiksen JH, Rasmussen LJ, Poirier F, Croci DO, Sundblad V, Rabinovich GA, Cerliani JP. Galectin-7 reprograms skin carcinogenesis by fostering innate immune evasive programs. Cell Death Differ 2023; 30:906-921. [PMID: 36693903 PMCID: PMC10070502 DOI: 10.1038/s41418-022-01108-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 01/25/2023] Open
Abstract
Non-melanoma skin cancer (NMSC) has risen dramatically as a result of chronic exposure to sunlight ultraviolet (UV) radiation, climatic changes and clinical conditions associated with immunosuppression. In spite of considerable progress, our understanding of the mechanisms that control NMSC development and their associated molecular and immunological landscapes is still limited. Here we demonstrated a critical role for galectin-7 (Gal-7), a β-galactoside-binding protein preferentially expressed in skin tissue, during NMSC development. Transgenic mice (Tg46) overexpressing Gal-7 in keratinocytes showed higher number of papillomas compared to WT mice or mice lacking Gal-7 (Lgals7-/-) when subjected to a skin carcinogenesis protocol, in which tumor initiator 7,12-dimethylbenz[a]anthracene (DMBA) and tumor promoter 12-O-tetradecanoyl-phorbol-13-acetate (TPA) were sequentially administered. RNAseq analysis of Tg46 tumor lesions revealed a unique profile compatible with cells of the myelomonocytic lineage infiltrating these tumors, an effect that was substantiated by a higher number of CD11b+Gr1+ cells in tumor-draining lymph nodes. Heightened c-Met activation and Cxcl-1 expression in Tg46 lesions suggested a contribution of this pathway to the recruitment of these cells. Remarkably, Gal-7 bound to the surface of CD11b+Ly6ChiLy6Glo monocytic myeloid cells and enhanced their immunosuppressive activity, as evidenced by increased IL-10 and TGF-β1 secretion, and higher T-cell inhibitory activity. In vivo, carcinogen-treated Lgals7-/- animals adoptively transferred with Gal-7-conditioned monocytic myeloid cells developed higher number of papillomas, whereas depletion of these cells in Tg46-treated mice led to reduction in the number of tumors. Finally, human NMSC biopsies showed increased LGALS7 mRNA and Gal-7 protein expression and displayed transcriptional profiles associated with myeloid programs, accompanied by elevated CXCL1 expression and c-Met activation. Thus, Gal-7 emerges as a critical mediator of skin carcinogenesis and a potential therapeutic target in human NMSC.
Collapse
Affiliation(s)
- Nicolás A Pinto
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Martín C Abba
- Centro de Investigaciones Inmunológicas Básicas y Aplicadas (CINIBA), Facultad de Ciencias Médicas, Universidad Nacional de La Plata, C1900, La Plata, Argentina
| | - Lorena Laporte
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Juan M Pérez Sáez
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Ada G Blidner
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Nicolás I Torres
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Rosa M Morales
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Sabrina G Gatto
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Camila A Bach
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| | - Florencia Veigas
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
- Universidad Argentina de la Empresa (UADE). Instituto de Tecnología (INTEC), C1073, Buenos Aires, Argentina
| | | | - Peng Song
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-, 2200, Copenhagen, Denmark
| | - Jane H Frederiksen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-, 2200, Copenhagen, Denmark
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-, 2200, Copenhagen, Denmark
| | - Francoise Poirier
- Institut Jacques Monod, UMR CNRS 7592, Paris-Diderot University, Paris, France
| | - Diego O Croci
- Instituto de Histología y Embriología de Mendoza Dr. Mario H. Burgos (IHEM-CONICET), Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, C5500, Mendoza, Argentina
| | - Victoria Sundblad
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina.
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina.
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428, Buenos Aires, Argentina.
| | - Juan P Cerliani
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428, Buenos Aires, Argentina
| |
Collapse
|
4
|
Roles of N-linked glycosylation and glycan-binding proteins in placentation: trophoblast infiltration, immunomodulation, angiogenesis, and pathophysiology. Biochem Soc Trans 2023; 51:639-653. [PMID: 36929183 DOI: 10.1042/bst20221406] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/18/2023]
Abstract
Protein N-linked glycosylation is a structurally diverse post-translational modification that stores biological information in a larger order of magnitude than other post-translational modifications such as phosphorylation, ubiquitination and acetylation. This gives N-glycosylated proteins a diverse range of properties and allows glyco-codes (glycan-related information) to be deciphered by glycan-binding proteins (GBPs). The intervillous space of the placenta is richly populated with membrane-bound and secreted glycoproteins. Evidence exists to suggest that altering the structural nature of their N-glycans can impact several trophoblast functions, which include those related to interactions with decidual cells. This review summarizes trophoblast-related activities influenced by N-glycan-GBP recognition, exploring how different subtypes of trophoblasts actively adapt to characteristics of the decidualized endometrium through cell-specific expression of N-glycosylated proteins, and how these cells receive decidua-derived signals via N-glycan-GBP interactions. We highlight work on how changes in N-glycosylation relates to the success of trophoblast infiltration, interactions of immunomodulators, and uterine angiogenesis. We also discuss studies that suggest aberrant N-glycosylation of trophoblasts may contribute to the pathogenesis of pregnancy complications (e.g. pre-eclampsia, early spontaneous miscarriages and hydatidiform mole). We propose that a more in-depth understanding of how N-glycosylation shapes trophoblast phenotype during early pregnancy has the potential to improve our approach to predicting, diagnosing and alleviating poor maternal/fetal outcomes associated with placental dysfunction.
Collapse
|
5
|
Santoprete R, Hourblin V, Foucher A, Dufour O, Bernard D, Domanov Y, Querleux B, Potter A. Reduction of wrinkles: From a computational hypothesis to a clinical, instrumental, and biological proof. Skin Res Technol 2023; 29:e13267. [PMID: 36973988 PMCID: PMC10155799 DOI: 10.1111/srt.13267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 12/06/2022] [Indexed: 03/09/2023]
Abstract
BACKGROUND Facial wrinkles are clear markers of the aging process, being chronological, photo-induced, or reflecting repetitive facial expressions. The aim of this study is to provide new insights into the biophysical and biological mechanisms involved in the formation, prevention, or elimination of the expression wrinkles. MATERIALS AND METHODS We use a computational model to get a better understanding of the wrinkle mechanical behavior and evolution after skin softening and suggesting a possible antiaging mechanism. Then, we provide a clinical demonstration of the anti-wrinkle effect of a long-term application of a 20% glycerol in a moisturizer formula (GBM) versus its vehicle on crow's feet. Skin hydration, elasticity, and wrinkles visibility were evaluated by a combination of clinical and instrumental in vivo data, inverse finite element analysis, and proteomic data. RESULTS The computational model shows a predominantly compressive stress beneath the wrinkle and its significant decrease by the softening of stratum corneum. The associated clinical study confirmed a significant increase of skin hydration and elasticity as well as a decrease of wrinkle visibility after 2 and 4 months as application for both formulas; this effect being stronger for GBM. A softening effect on stratum corneum and dermis was also observed for the GBM. Furthermore, proteomic data revealed an effect of upregulation of four proteins associated with desquamation, cell-glycan extracellular interactions, and protein glycation/oxidation, functions related to the tissue mechanics and adhesion. CONCLUSIONS We provide an in vivo demonstration of the anti-ageing benefit of glycerol at high dose (20%) reflected by a cumulative skin surface softening effect. The use of high moisturizing potent formulations should bring additional performance to other conventional moisturizing formulations.
Collapse
Affiliation(s)
| | | | - Aude Foucher
- L'Oréal Research & InnovationAulnay‐sous‐BoisFrance
| | | | | | | | | | - Anne Potter
- L'Oréal Research & InnovationAulnay‐sous‐BoisFrance
| |
Collapse
|
6
|
Kruk L, Braun A, Cosset E, Gudermann T, Mammadova-Bach E. Galectin functions in cancer-associated inflammation and thrombosis. Front Cardiovasc Med 2023; 10:1052959. [PMID: 36873388 PMCID: PMC9981828 DOI: 10.3389/fcvm.2023.1052959] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 01/12/2023] [Indexed: 02/19/2023] Open
Abstract
Galectins are carbohydrate-binding proteins that regulate many cellular functions including proliferation, adhesion, migration, and phagocytosis. Increasing experimental and clinical evidence indicates that galectins influence many steps of cancer development by inducing the recruitment of immune cells to the inflammatory sites and modulating the effector function of neutrophils, monocytes, and lymphocytes. Recent studies described that different isoforms of galectins can induce platelet adhesion, aggregation, and granule release through the interaction with platelet-specific glycoproteins and integrins. Patients with cancer and/or deep-venous thrombosis have increased levels of galectins in the vasculature, suggesting that these proteins could be important contributors to cancer-associated inflammation and thrombosis. In this review, we summarize the pathological role of galectins in inflammatory and thrombotic events, influencing tumor progression and metastasis. We also discuss the potential of anti-cancer therapies targeting galectins in the pathological context of cancer-associated inflammation and thrombosis.
Collapse
Affiliation(s)
- Linus Kruk
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany.,Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Attila Braun
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Erika Cosset
- CRCL, UMR INSERM 1052, CNRS 5286, Centre Léon Bérard, Lyon, France
| | - Thomas Gudermann
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany.,German Center for Lung Research (DZL), Munich, Germany
| | - Elmina Mammadova-Bach
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany.,Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Munich, Germany
| |
Collapse
|
7
|
Čoma M, Manning JC, Kaltner H, Gál P. The sweet side of wound healing: galectins as promising therapeutic targets in hemostasis, inflammation, proliferation, and maturation/remodeling. Expert Opin Ther Targets 2023; 27:41-53. [PMID: 36716023 DOI: 10.1080/14728222.2023.2175318] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Understanding the molecular and cellular processes involved in skin wound healing may pave the way for the development of innovative approaches to transforming the identified natural effectors into therapeutic tools. Based on the extensive involvement of the ga(lactoside-binding)lectin family in (patho)physiological processes, it has been well established that galectins are involved in a wide range of cell-cell and cell-matrix interactions. AREAS COVERED In the present paper, we provide an overview of the biological role of galectins in repair and regeneration, focusing on four main phases (hemostasis, inflammation, proliferation, and maturation/remodeling) of skin repair using basic wound models (open excision vs. sutured incision). EXPERT OPINION The reported data make a strong case for directing further efforts to treat excisional and incisional wounds differently. Functions of galectins essentially result from their modular presentation. In fact, Gal-1 seems to play a role in the early phases of healing (anti-inflammatory) and wound contraction, Gal-3 accelerates re-epithelization and increases tensile strength (scar inductor). Galectins have also become subject of redesigning by engineering to optimize the activity. Clinically relevant, these new tools derived from the carbohydrate recognition domain platform may also prove helpful for other purposes, such as potent antibacterial agglutinins and opsonins.
Collapse
Affiliation(s)
- Matúš Čoma
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases Inc, Košice, Slovak Republic.,Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Joachim C Manning
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilian University, Munich, Germany
| | - Herbert Kaltner
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilian University, Munich, Germany
| | - Peter Gál
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases Inc, Košice, Slovak Republic.,Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic.,Prague Burn Center, Third Faculty of Medicine, Charles University and University Hospital Královske Vinohrady, Prague, Czech Republic.,Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University, Bratislava, Slovak Republic
| |
Collapse
|
8
|
Cárdenas-León CG, Mäemets-Allas K, Klaas M, Lagus H, Kankuri E, Jaks V. Matricellular proteins in cutaneous wound healing. Front Cell Dev Biol 2022; 10:1073320. [PMID: 36506087 PMCID: PMC9730256 DOI: 10.3389/fcell.2022.1073320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Cutaneous wound healing is a complex process that encompasses alterations in all aspects of the skin including the extracellular matrix (ECM). ECM consist of large structural proteins such as collagens and elastin as well as smaller proteins with mainly regulative properties called matricellular proteins. Matricellular proteins bind to structural proteins and their functions include but are not limited to interaction with cell surface receptors, cytokines, or protease and evoking a cellular response. The signaling initiated by matricellular proteins modulates differentiation and proliferation of cells having an impact on the tissue regeneration. In this review we give an overview of the matricellular proteins that have been found to be involved in cutaneous wound healing and summarize the information known to date about their functions in this process.
Collapse
Affiliation(s)
| | - Kristina Mäemets-Allas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Mariliis Klaas
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Heli Lagus
- Department of Plastic Surgery and Wound Healing Centre, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Viljar Jaks
- Department of Cell Biology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia,Dermatology Clinic, Tartu University Clinics, Tartu, Estonia,*Correspondence: Viljar Jaks,
| |
Collapse
|
9
|
Yu D, Bu M, Yu P, Li Y, Chong Y. Regulation of wound healing and fibrosis by galectins. J Mol Med (Berl) 2022; 100:861-874. [PMID: 35589840 DOI: 10.1007/s00109-022-02207-1] [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: 12/02/2021] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 11/28/2022]
Abstract
Galectins are a family of proteins with at least one carbohydrate-recognition domain. Galectins are present in various tissues and organs and participate in different physiological and pathological molecular reactions in vivo. Wound healing is the basic process of traumatic disease recovery. Wound healing involves three overlapping stages: inflammation, proliferation, and remodelling. Furthermore, a comparison of wound healing with the tumour microenvironment revealed that galectin plays a key role in the wound healing process. The current review describes the role of galectin in inflammation, angiogenesis, re-epithelialisation, and fibrous scar formation and evaluates its potential as a therapeutic drug for wounds.
Collapse
Affiliation(s)
- Dong Yu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China.,Department of Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Ming Bu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China.,Department of Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Ping Yu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Yaping Li
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China.,Department of Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Yang Chong
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, Jiangsu, China. .,Department of Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu, China.
| |
Collapse
|
10
|
Verkerke H, Dias-Baruffi M, Cummings RD, Arthur CM, Stowell SR. Galectins: An Ancient Family of Carbohydrate Binding Proteins with Modern Functions. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2442:1-40. [PMID: 35320517 DOI: 10.1007/978-1-0716-2055-7_1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Galectins are a large family of carbohydrate binding proteins with members in nearly every lineage of multicellular life. Through tandem and en-mass genome duplications, over 15 known vertebrate galectins likely evolved from a single common ancestor extant in pre-chordate lineages. While galectins have divergently evolved numerous functions, some of which do not involve carbohydrate recognition, the vast majority of the galectins have retained the conserved ability to bind variably modified polylactosamine (polyLacNAc) residues on glycans that modify proteins and lipids on the surface of host cells and pathogens. In addition to their direct role in microbial killing, many proposed galectin functions in the immune system and cancer involve crosslinking glycosylated receptors and modifying signaling pathways or sensitivity to antigen from the outside in. However, a large body of work has uncovered intracellular galectin functions mediated by carbohydrate- and non-carbohydrate-dependent interactions. In the cytoplasm, galectins can tune intracellular kinase and G-protein-coupled signaling cascades important for nutrient sensing, cell cycle progression, and transformation. Particularly, but interconnected pathways, cytoplasmic galectins serve the innate immune system as sensors of endolysosomal damage, recruiting and assembling the components of autophagosomes during intracellular infection through carbohydrate-dependent and -independent activities. In the nucleus, galectins participate in pre-mRNA splicing perhaps through interactions with non-coding RNAs required for assembly of spliceosomes. Together, studies of galectin function paint a picture of a functionally dynamic protein family recruited during eons of evolution to regulate numerous essential cellular processes in the context of multicellular life.
Collapse
Affiliation(s)
- Hans Verkerke
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA
| | - Marcelo Dias-Baruffi
- Department of Clinical Analysis, Toxicological and Bromatological, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Connie M Arthur
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Harvard Glycomics Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Menkhorst E, Than NG, Jeschke U, Barrientos G, Szereday L, Dveksler G, Blois SM. Medawar's PostEra: Galectins Emerged as Key Players During Fetal-Maternal Glycoimmune Adaptation. Front Immunol 2022; 12:784473. [PMID: 34975875 PMCID: PMC8715898 DOI: 10.3389/fimmu.2021.784473] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Lectin-glycan interactions, in particular those mediated by the galectin family, regulate many processes required for a successful pregnancy. Over the past decades, increasing evidence gathered from in vitro and in vivo experiments indicate that members of the galectin family specifically bind to both intracellular and membrane bound carbohydrate ligands regulating angiogenesis, immune-cell adaptations required to tolerate the fetal semi-allograft and mammalian embryogenesis. Therefore, galectins play important roles in fetal development and placentation contributing to maternal and fetal health. This review discusses the expression and role of galectins during the course of pregnancy, with an emphasis on maternal immune adaptions and galectin-glycan interactions uncovered in the recent years. In addition, we summarize the galectin fingerprints associated with pathological gestation with particular focus on preeclampsia.
Collapse
Affiliation(s)
- Ellen Menkhorst
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia.,Gynaecological Research Centre, The Women's Hospital, Melbourne, VIC, Australia
| | - Nandor Gabor Than
- Systems Biology of Reproduction Research Group, Institute of Enyzmology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital Augsburg, Augsburg, Germany
| | - Gabriela Barrientos
- Laboratorio de Medicina Experimental, Hospital Alemán-Consejo Nacional de Investigaciones Científicas y Técnicas, Ciudad Autónoma de Buenos Aires, Argentina
| | - Laszlo Szereday
- Medical School, Department of Medical Microbiology and Immunology, University of Pecs, Pecs, Hungary
| | - Gabriela Dveksler
- Department of Pathology, Uniformed Services University, Bethesda, MD, United States
| | - Sandra M Blois
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
12
|
Cerri DG, Rodrigues LC, Fermino ML, Papoti M, Cummings RD, Stowell SR, Dias-Baruffi M. Investigation of Galectins in Frozen Tissue and Mammalian Cell Culture Using Confocal Miccroscopy. Methods Mol Biol 2022; 2442:289-306. [PMID: 35320532 DOI: 10.1007/978-1-0716-2055-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Galectins are multifunctional glycan-binding proteins present in various tissues that participate in multiple physiological and pathological processes and are considered as not only biomarkers of human diseases but also molecular targets for treating cancer and inflammatory illnesses in many organs. In the glycobiology field, it is crucial to determine the pattern of galectin expression and location in cells and tissues. Confocal microscopy is a powerful imaging technology that represents a unique approach to investigate the expression and location of biomolecules in various tissues and cells. The confocal microscope acquires images of the specimen through the reflected or fluorescent light from the objective's focal plane, using laser light focused on a small spot inside the tissue or cell. This technique provides high-resolution and high-contrast images without artifacts generated by conventional microscopy and enables reconstruction of virtual tridimensional images by acquiring multiple sections from several focal planes, which makes it possible to obtain the precise spatial location of any cellular structure or molecule. Furthermore, confocal microscopy is a non-invasive tissue imaging strategy used in clinical practices. We describe herein the immunofluorescence confocal method for examining galectins in frozen tissue sections and mammalian cell culture.
Collapse
Affiliation(s)
- Daniel Giuliano Cerri
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacênuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Lilian Cataldi Rodrigues
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacênuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Marise Lopes Fermino
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacênuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Marcelo Papoti
- Escola de Educação Física e Esporte de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Richard D Cummings
- Department of Surgery, National Center for Functional Glycomics, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sean R Stowell
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Marcelo Dias-Baruffi
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas da Faculdade de Ciências Farmacênuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil.
| |
Collapse
|
13
|
The Immunogenetics of Morphea and Lichen Sclerosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:155-172. [DOI: 10.1007/978-3-030-92616-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
14
|
Proux-Gillardeaux V, Advedissian T, Perin C, Gelly JC, Viguier M, Deshayes F. Identification of a new regulation pathway of EGFR and E-cadherin dynamics. Sci Rep 2021; 11:22705. [PMID: 34811416 PMCID: PMC8609017 DOI: 10.1038/s41598-021-02042-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022] Open
Abstract
E-cadherin and EGFR are known to be closely associated hence regulating differentiation and proliferation notably in epithelia. We have previously shown that galectin-7 binds to E-cadherin and favors its retention at the plasma membrane. In this study, we shed in light that galectin-7 establishes a physical link between E-cadherin and EGFR. Indeed, our results demonstrate that galectin-7 also binds to EGFR, but unlike the binding to E-cadherin this binding is sugar dependent. The establishment of E-cadherin/EGFR complex and the binding of galectin-7 to EGFR thus lead to a regulation of its signaling and intracellular trafficking allowing cell proliferation and migration control. In vivo observations further support these results since an epidermal thickening is observed in galectin-7 deficient mice. This study therefore reveals that galectin-7 controls epidermal homeostasis through the regulation of E-cadherin/EGFR balance.
Collapse
Affiliation(s)
| | - Tamara Advedissian
- Membrane Traffic and Cell Division Laboratory, Institut Pasteur, UMR3691, CNRS, 75015, Paris, France
| | - Charlotte Perin
- Université de Paris, UMR_S1134, BIGR, Inserm, 75006, Paris, France.,Institut National de Transfusion Sanguine, 75015, Paris, France
| | - Jean-Christophe Gelly
- Université de Paris, UMR_S1134, BIGR, Inserm, 75006, Paris, France.,Institut National de Transfusion Sanguine, 75015, Paris, France
| | - Mireille Viguier
- CNRS, Institut Jacques Monod, Université de Paris, F-75013, Paris, France
| | | |
Collapse
|
15
|
Sewgobind NV, Albers S, Pieters RJ. Functions and Inhibition of Galectin-7, an Emerging Target in Cellular Pathophysiology. Biomolecules 2021; 11:1720. [PMID: 34827718 PMCID: PMC8615947 DOI: 10.3390/biom11111720] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 12/16/2022] Open
Abstract
Galectin-7 is a soluble unglycosylated lectin that is able to bind specifically to β-galactosides. It has been described to be involved in apoptosis, proliferation and differentiation, but also in cell adhesion and migration. Several disorders and diseases are discussed by covering the aforementioned biological processes. Structural features of galectin-7 are discussed as well as targeting the protein intracellularly or extracellularly. The exact molecular mechanisms that lie behind many biological processes involving galectin-7 are not known. It is therefore useful to come up with chemical probes or tools in order to obtain knowledge of the physiological processes. The objective of this review is to summarize the roles and functions of galectin-7 in the human body, providing reasons why it is necessary to design inhibitors for galectin-7, to give the reader structural insights and describe its current inhibitors.
Collapse
Affiliation(s)
| | | | - Roland J. Pieters
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, P.O. Box 80082, NL-3508 TB Utrecht, The Netherlands; (N.V.S.); (S.A.)
| |
Collapse
|
16
|
Girousi E, Muerner L, Parisi L, Rihs S, von Gunten S, Katsaros C, Degen M. Lack of IRF6 Disrupts Human Epithelial Homeostasis by Altering Colony Morphology, Migration Pattern, and Differentiation Potential of Keratinocytes. Front Cell Dev Biol 2021; 9:718066. [PMID: 34660580 PMCID: PMC8514984 DOI: 10.3389/fcell.2021.718066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/16/2021] [Indexed: 12/03/2022] Open
Abstract
Variants within the gene encoding for the transcription factor Interferon Regulatory Factor 6 (IRF6) are associated with syndromic and non-syndromic Cleft Lip/Palate (CLP) cases. IRF6 plays a vital role in the regulation of the proliferation/differentiation balance in keratinocytes and is involved in wound healing and migration. Since a fraction of CLP patients undergoing corrective cleft surgery experience wound healing complications, IRF6 represents an interesting candidate gene linking the two processes. However, Irf6 function has been mainly studied in mice and knowledge on IRF6 in human cells remains sparse. Here, we aimed to elucidate the role of IRF6 in human postnatal skin- and oral mucosa-derived keratinocytes. To do so, we applied CRISPR/Cas9 to ablate IRF6 in two TERT-immortalized keratinocyte cultures, which we used as model cell lines. We show that IRF6 controls the appearance of single cells and colonies, with the latter being less cohesive in its absence. Consequently, IRF6 knockout keratinocytes often moved as single cells instead of a collective epithelial sheet migration but maintained their epithelial character. Lack of IRF6 triggered severe keratinocyte differentiation defects, which were already apparent in the stratum spinosum and extended to the stratum corneum in 3D organotypic skin cultures, while it did not alter their growth rate. Finally, proteomics revealed that most of the differentially expressed proteins in the absence of IRF6 could be associated with differentiation, cell-cell adhesion as well as immune response. Our data expand the knowledge on IRF6 in human postnatal keratinocytes, which will help to better understand IRF6-related pathologies.
Collapse
Affiliation(s)
- Eleftheria Girousi
- Laboratory for Oral Molecular Biology, Dental Research Center, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | - Lukas Muerner
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Ludovica Parisi
- Laboratory for Oral Molecular Biology, Dental Research Center, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | - Silvia Rihs
- Laboratory for Oral Molecular Biology, Dental Research Center, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | | | - Christos Katsaros
- Laboratory for Oral Molecular Biology, Dental Research Center, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | - Martin Degen
- Laboratory for Oral Molecular Biology, Dental Research Center, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| |
Collapse
|
17
|
Pinto NA, Rabinovich GA, Sundblad V, Cerliani JP. Enhanced galectin-7 expression favors wound healing. J Dermatol 2021; 48:1616-1618. [PMID: 34318956 DOI: 10.1111/1346-8138.16048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/10/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Nicolás A Pinto
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Victoria Sundblad
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Juan P Cerliani
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
18
|
Maruszewska-Cheruiyot M, Stear M, Donskow-Łysoniewska K. Galectins - Important players of the immune response to CNS parasitic infection. Brain Behav Immun Health 2021; 13:100221. [PMID: 34589740 PMCID: PMC8474370 DOI: 10.1016/j.bbih.2021.100221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/18/2021] [Accepted: 01/30/2021] [Indexed: 11/18/2022] Open
Abstract
Galectins are a family of proteins that bind β-galactosides and play key roles in a variety of cellular processes including host defense and entry of parasites into the host cells. They have been well studied in hosts but less so in parasites. As both host and parasite galectins are highly upregulated proteins following infection, galectins are an area of increasing interest and their role in immune modulation has only recently become clear. Correlation of CNS parasitic diseases with mental disorders as a result of direct or indirect interaction has been observed. Therefore, galectins produced by the parasite should be taken into consideration as potential therapeutic agents.
Collapse
Affiliation(s)
- Marta Maruszewska-Cheruiyot
- Laboratory of Parasitology, General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163, Warsaw, Poland
- Corresponding author.
| | - Michael Stear
- Department of Animal, Plant and Soil Science, Agribio, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Katarzyna Donskow-Łysoniewska
- Laboratory of Parasitology, General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163, Warsaw, Poland
| |
Collapse
|
19
|
Fujimoto N, Akiyama M, Satoh Y, Tajima S. Interaction of galectin-7 with HMGCS1 in vitro may facilitate cholesterol deposition in cultured keratinocytes. J Invest Dermatol 2021; 142:539-548. [PMID: 34454908 DOI: 10.1016/j.jid.2021.04.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 03/15/2021] [Accepted: 04/07/2021] [Indexed: 01/20/2023]
Abstract
Three-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) synthase 1 (HMGCS1) was identified to interact with Gal-7, a pro-apoptotic β-galactoside binding protein, by yeast two-hybrid system. Their interaction was confirmed by in vitro β-galactosidase, BIA core and immunoprecipitation assays. Distinct interactive site of HMGCS1was found to reside at Phe-26. The expression of HMGCS1 in cultured keratinocytes was up-regulated by exogenous Gal-7 and down-regulated in Gal-7 siRNA transfected cells. HMGCS1-overexpressing cells were found to induce Gal-7 expression, which suggests that Gal-7 and HMGCS1 expressions are both stimulated by a positive feedback regulation. The amount of cholesterol, a final biosynthetic product of HMGCS1-involved pathway, was increased in Gal-7 treated cells, and was significantly reduced in Gal-7 siRNA transfected cells. The increase of cholesterol level in Gal-7 treated cells was inhibited by wild type HMGCS1 peptide but not by Phe-26 mutated peptide, suggesting that the interaction of Gal-7/HMGCS1 is related to cellular cholesterol level. Foam cells in granulomatous tissues of the specimens from normolipemic cutaneous xanthoma showed positive reactions with the antibodies for Gal-7 and HMGCS1 as well as lipid markers. These results are likely to indicate that Gal-7 induction in epidermal keratinocytes causes both apoptotic cell death and HMGCS1-mediated cholesterol accumulation which will be phagocytized by macrophages. This mechanism may explain the pathogenesis of normolipemic cutaneous xanthoma.
Collapse
Affiliation(s)
- Norihiro Fujimoto
- Departments of Dermatology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Minoru Akiyama
- Departments of Dermatology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Yasushi Satoh
- Departments of Biochemistry, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Shingo Tajima
- Departments of Dermatology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| |
Collapse
|
20
|
Huang CY, Hsieh PL, Ng MY, Liao YW, Yu CC, Lin T. Galectin-7 promotes proliferation and wound healing capacities in periodontal ligament fibroblasts by activating ERK signaling. J Formos Med Assoc 2021; 121:1008-1011. [PMID: 34452782 DOI: 10.1016/j.jfma.2021.08.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/29/2021] [Accepted: 08/11/2021] [Indexed: 11/26/2022] Open
Abstract
Periodontitis is a progressive inflammation condition and a primary cause of tooth loss in adults. As one of the abundant cell types in the periodontium, periodontal ligament fibroblasts (PDLFs) play an integral role in the maintenance and regeneration of periodontal tissue. Our previous work has shown that the application of Er:YAG laser increased the cell proliferation and migratory capacity of PDLFs via induction of galectin-7. In the present study, we aimed to evaluate if the forced expression of galectin-7 directly affected the cellular phenotypes of PDLFs. Our results showed that the cell proliferation, transwell migration, invasion, and wound healing capacities were all upregulated in PDLFs with the ectopic expression of galectin-7. These results suggest that therapeutic approaches to enhance the expression of galectin-7 in periodontium may accelerate tissue regeneration by recruiting more PDLFs to the injured site.
Collapse
Affiliation(s)
- Chao-Yen Huang
- Department of Emergency Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Min Yee Ng
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Wen Liao
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan; Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Taichen Lin
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
21
|
Imitating evolution's tinkering by protein engineering reveals extension of human galectin-7 activity. Histochem Cell Biol 2021; 156:253-272. [PMID: 34152508 PMCID: PMC8460509 DOI: 10.1007/s00418-021-02004-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2021] [Indexed: 12/23/2022]
Abstract
Wild-type lectins have distinct types of modular design. As a step to explain the physiological importance of their special status, hypothesis-driven protein engineering is used to generate variants. Concerning adhesion/growth-regulatory galectins, non-covalently associated homodimers are commonly encountered in vertebrates. The homodimeric galectin-7 (Gal-7) is a multifunctional context-dependent modulator. Since the possibility of conversion from the homodimer to hybrids with other galectin domains, i.e. from Gal-1 and Gal-3, has recently been discovered, we designed Gal-7-based constructs, i.e. stable (covalently linked) homo- and heterodimers. They were produced and purified by affinity chromatography, and the sugar-binding activity of each lectin unit proven by calorimetry. Inspection of profiles of binding of labeled galectins to an array-like platform with various cell types, i.e. sections of murine epididymis and jejunum, and impact on neuroblastoma cell proliferation revealed no major difference between natural and artificial (stable) homodimers. When analyzing heterodimers, acquisition of altered properties was seen. Remarkably, binding properties and activity as effector can depend on the order of arrangement of lectin domains (from N- to C-termini) and on the linker length. After dissociation of the homodimer, the Gal-7 domain can build new functionally active hybrids with other partners. This study provides a clear direction for research on defining the full range of Gal-7 functionality and offers the perspective of testing applications for engineered heterodimers.
Collapse
|
22
|
Jeethy Ram T, Lekshmi A, Somanathan T, Sujathan K. Galectin-3: A factotum in carcinogenesis bestowing an archery for prevention. Tumour Biol 2021; 43:77-96. [PMID: 33998569 DOI: 10.3233/tub-200051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Cancer metastasis and therapy resistance are the foremost hurdles in oncology at the moment. This review aims to pinpoint the functional aspects of a unique multifaceted glycosylated molecule in both intracellular and extracellular compartments of a cell namely galectin-3 along with its metastatic potential in different types of cancer. All materials reviewed here were collected through the search engines PubMed, Scopus, and Google scholar. Among the 15 galectins identified, the chimeric gal-3 plays an indispensable role in the differentiation, transformation, and multi-step process of tumor metastasis. It has been implicated in the molecular mechanisms that allow the cancer cells to survive in the intravascular milieu and promote tumor cell extravasation, ultimately leading to metastasis. Gal-3 has also been found to have a pivotal role in immune surveillance and pro-angiogenesis and several studies have pointed out the importance of gal-3 in establishing a resistant phenotype, particularly through the epithelial-mesenchymal transition process. Additionally, some recent findings suggest the use of gal-3 inhibitors in overcoming therapeutic resistance. All these reports suggest that the deregulation of these specific lectins at the cellular level could inhibit cancer progression and metastasis. A more systematic study of glycosylation in clinical samples along with the development of selective gal-3 antagonists inhibiting the activity of these molecules at the cellular level offers an innovative strategy for primary cancer prevention.
Collapse
Affiliation(s)
- T Jeethy Ram
- Division of Cancer Research, Regional Cancer Centre, Medical College, Trivandrum, Kerala, India
| | - Asha Lekshmi
- Division of Cancer Research, Regional Cancer Centre, Medical College, Trivandrum, Kerala, India
| | - Thara Somanathan
- Division of Pathology, Regional Cancer Centre, Medical College, Trivandrum, Kerala, India
| | - K Sujathan
- Regional Cancer Centre, Thiruvananthapuram, Kerala, India
| |
Collapse
|
23
|
Kim S, Kim Y, Hyun YS, Choi H, Kim SY, Kim TG. Exosomes from human cord blood plasma accelerate cutaneous wound healing by promoting fibroblast function, angiogenesis, and M2 macrophage differentiation. Biomater Sci 2021; 9:3028-3039. [PMID: 33657200 DOI: 10.1039/d0bm01801e] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Exosomes contain natural cargo molecules, such as miRNA, mRNA, and proteins, and transfer these functional cargos to neighboring or distant cells through circulation. In the wound-healing process, exosomes in the human blood and body fluids perform various functions, including proliferation, angiogenesis, differentiation, and wound healing, owing to their unique compositions. However, there is very limited information on the wound-healing effect of proteins in human cord blood plasma exosomes (CBPexo). Therefore, we studied the wound-healing potential of these proteins in terms of fibroblast functions, angiogenesis, and M2 macrophage differentiation. When scratch wound assays were conducted using human fibroblasts, CBPexo exhibited better wound-healing effects than adult blood plasma exosomes (ABPexo). CBPexo also promoted angiogenesis and differentiation of M2 macrophages, thus promoting the transition from inflammation to proliferation. To evaluate the CBPexo molecules involved, five proteins, GAL-3, GAL-7, HSP-72, PIP, and S100-A7, were selected through proteomic analysis, and their functions were investigated using an artificial exosome that expresses these proteins. Among these, HSP72 and PIP exhibited wound-healing effects similar to CBPexo. Furthermore, artificial exosomes expressing both HSP72 and PIP showed better wound-healing effects than CBPexo. Therefore, the use of artificial CBPexo can potentially overcome the limitations related to exosome production from CB.
Collapse
Affiliation(s)
- Sueon Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. and Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yeongwon Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. and Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - You-Seok Hyun
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. and Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Haeyoun Choi
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. and Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Su-Yeon Kim
- Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Tai-Gyu Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. and Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea and Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
24
|
Tian J, He R, Fan Y, Zhang Q, Tian B, Zhou C, Liu C, Song M, Zhao S. Galectin-7 overexpression destroys airway epithelial barrier in transgenic mice. Integr Zool 2021; 16:270-279. [PMID: 32627954 DOI: 10.1111/1749-4877.12463] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
When the integrity of airway epithelium is destroyed, the ordered airway barrier no longer exists and increases sensitivity to viral infections and allergens, leading to the occurrence of airway inflammation such as asthma. Here, we found that galectin-7 transgenic(+) mice exhibited abnormal airway structures as embryos and after birth. These abnormalities included absent or substantially reduced pseudostratified columnar ciliated epithelium and increased monolayer cells with irregular arrangement and widening of intercellular spaces. Moreover, airway tissue from galectin-7 transgenic(+) mice showed evidence of impaired cell-cell junctions and decreased expression of zonula occludens-1(ZO-1) and E-cadherin. When treated with respiratory syncytial virus (RSV) or ovalbumin (OVA), galectin-7 transgenic(+) mice developed substantially increased bronchial epithelial detachment and apoptosis, airway smooth muscle and basement membrane thickening, and enhanced airway responsiveness. We found that Galectin-7 localized in the cytoplasm and nucleus of bronchial epithelial cells, and that increased apoptosis was mediated through mitochondrial release of cytochrome c and upregulated JNK1 activation and expression of caspase-3 in galectin-7 Tg(+) mice. These findings suggested that Galectin-7 causes airway structural defects and destroys airway epithelium barrier, which predispose the airways to RSV or OVA-induced epithelial apoptosis, injury, and other asthma responses.
Collapse
Affiliation(s)
- Jing Tian
- Department of Respiratory Medicine II, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Ruxuan He
- Department of Respiratory Medicine II, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yimu Fan
- Department of Respiratory Medicine II, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Qianqian Zhang
- Department of Respiratory Medicine II, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Baolin Tian
- Department of Respiratory Medicine II, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chunju Zhou
- Virus Laboratory, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Chunyan Liu
- Virus Laboratory, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, China
| | - Mingjing Song
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Beijing Engineering Research Center for Experimental Animal Models of Human Diseases, Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China
| | - Shunying Zhao
- Department of Respiratory Medicine II, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
25
|
Lin CY, Nozawa T, Minowa-Nozawa A, Toh H, Hikichi M, Iibushi J, Nakagawa I. Autophagy Receptor Tollip Facilitates Bacterial Autophagy by Recruiting Galectin-7 in Response to Group A Streptococcus Infection. Front Cell Infect Microbiol 2021; 10:583137. [PMID: 33425778 PMCID: PMC7786282 DOI: 10.3389/fcimb.2020.583137] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/19/2020] [Indexed: 01/09/2023] Open
Abstract
Bacterial autophagy—a type of macroautophagy that is also termed xenophagy—selectively targets intracellular bacteria such as group A Streptococcus (GAS), a ubiquitous pathogen that causes numerous serious diseases, including pharyngitis, skin infections, and invasive life-threatening infections. Although bacterial autophagy is known to eliminate invading bacteria via the action of autophagy receptors, the underlying mechanism remains unclear. Herein, we elucidated that Tollip functions as a bacterial-autophagy receptor in addition to participating involved in the intracellular immunity mechanism that defends against bacterial infection. Tollip was recruited to GAS-containing endosomal vacuoles prior to the escape of GAS into the cytosol; additionally, Tollip knockout disrupted the recruitment of other autophagy receptors, such as NBR1, TAX1BP1, and NDP52, to GAS-containing autophagosomes and led to prolonged intracellular survival of GAS. Furthermore, Tollip was found to be required for the recruitment of galectin-1 and -7 to GAS-containing autophagosomes, and immunoprecipitation results indicated that Tollip interacts with galectin-7. Lastly, our data also revealed that galectin-1 and -7 are involved in the restriction of GAS replication in cells. These results demonstrated that Tollip modulates bacterial autophagy by recruiting other autophagy receptors and galectins.
Collapse
Affiliation(s)
- Ching-Yu Lin
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsuko Minowa-Nozawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hirotaka Toh
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Miyako Hikichi
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junpei Iibushi
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ichiro Nakagawa
- Department of Microbiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
26
|
Chen HL, Lo CH, Huang CC, Lu MP, Hu PY, Chen CS, Chueh DY, Chen P, Lin TN, Lo YH, Hsiao YP, Hsu DK, Liu FT. Galectin-7 downregulation in lesional keratinocytes contributes to enhanced IL-17A signaling and skin pathology in psoriasis. J Clin Invest 2021; 131:130740. [PMID: 33055419 DOI: 10.1172/jci130740] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/02/2020] [Indexed: 12/22/2022] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by inflammatory cell infiltration, as well as hyperproliferation of keratinocytes in skin lesions, and is considered a metabolic syndrome. We found that the expression of galectin-7 is reduced in skin lesions of patients with psoriasis. IL-17A and TNF-α, 2 cytokines intimately involved in the development of psoriatic lesions, suppressed galectin-7 expression in human primary keratinocytes (HEKn cells) and the immortalized human keratinocyte cell line HaCaT. A galectin-7 knockdown in these cells elevated the production of IL-6 and IL-8 and enhanced ERK signaling when the cells were stimulated with IL-17A. Galectin-7 attenuated IL-17A-induced production of inflammatory mediators by keratinocytes via the microRNA-146a/ERK pathway. Moreover, galectin-7-deficient mice showed enhanced epidermal hyperplasia and skin inflammation in response to intradermal IL-23 injection. We identified fluvastatin as an inducer of galectin-7 expression by connectivity map analysis, confirmed this effect in keratinocytes, and demonstrated that fluvastatin attenuated IL-6 and IL-8 production induced by IL-17A. Thus, we validate a role of galectin-7 in the pathogenesis of psoriasis, in both epidermal hyperplasia and keratinocyte-mediated inflammatory responses, and formulate a rationale for the use of statins in the treatment of psoriasis.
Collapse
Affiliation(s)
- Hung-Lin Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chia-Hui Lo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chi-Chun Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Meng-Ping Lu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Po-Yuan Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chang-Shan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Di-Yen Chueh
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Peilin Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Teng-Nan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuan-Hsin Lo
- Department of Dermatology, Fu Jen Catholic University Hospital, and.,School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yu-Ping Hsiao
- Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Daniel K Hsu
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, USA
| |
Collapse
|
27
|
Lin T, Yu CC, Liu CM, Hsieh PL, Liao YW, Yu CH, Chen CJ. Er:YAG laser promotes proliferation and wound healing capacity of human periodontal ligament fibroblasts through Galectin-7 induction. J Formos Med Assoc 2021; 120:388-394. [DOI: 10.1016/j.jfma.2020.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/21/2020] [Accepted: 06/03/2020] [Indexed: 12/16/2022] Open
|
28
|
Girotti MR, Salatino M, Dalotto-Moreno T, Rabinovich GA. Sweetening the hallmarks of cancer: Galectins as multifunctional mediators of tumor progression. J Exp Med 2020; 217:133540. [PMID: 31873723 PMCID: PMC7041721 DOI: 10.1084/jem.20182041] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/14/2019] [Accepted: 11/18/2019] [Indexed: 12/25/2022] Open
Abstract
Hanahan and Weinberg have proposed 10 organizing principles that enable growth and metastatic dissemination of cancer cells. These distinctive and complementary capabilities, defined as the "hallmarks of cancer," include the ability of tumor cells and their microenvironment to sustain proliferative signaling, evade growth suppressors, resist cell death, promote replicative immortality, induce angiogenesis, support invasion and metastasis, reprogram energy metabolism, induce genomic instability and inflammation, and trigger evasion of immune responses. These common features are hierarchically regulated through different mechanisms, including those involving glycosylation-dependent programs that influence the biological and clinical impact of each hallmark. Galectins, an evolutionarily conserved family of glycan-binding proteins, have broad influence in tumor progression by rewiring intracellular and extracellular circuits either in cancer or stromal cells, including immune cells, endothelial cells, and fibroblasts. In this review, we dissect the role of galectins in shaping cellular circuitries governing each hallmark of tumors, illustrating relevant examples and highlighting novel opportunities for treating human cancer.
Collapse
Affiliation(s)
- María Romina Girotti
- Laboratorio de Inmuno-Oncología Traslacional, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Mariana Salatino
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Tomás Dalotto-Moreno
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
29
|
Menkhorst E, Zhou W, Santos LL, Delforce S, So T, Rainczuk K, Loke H, Syngelaki A, Varshney S, Williamson N, Pringle K, Young MJ, Nicolaides KH, St-Pierre Y, Dimitriadis E. Galectin-7 Impairs Placentation and Causes Preeclampsia Features in Mice. Hypertension 2020; 76:1185-1194. [PMID: 32862708 DOI: 10.1161/hypertensionaha.120.15313] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Preeclampsia is a serious pregnancy-induced disorder unique to humans. The etiology of preeclampsia is poorly understood; however, poor placental formation is thought causal. Galectin-7 is produced by trophoblast and is elevated in first-trimester serum of women who subsequently develop preeclampsia. We hypothesized that elevated placental galectin-7 may be causative of preeclampsia. Here, we demonstrated increased galectin-7 production in chorionic villous samples from women who subsequently develop preterm preeclampsia compared with uncomplicated pregnancies. In vitro, galectin-7 impaired human first-trimester trophoblast outgrowth, increased placental production of the antiangiogenic sFlt-1 splice variant, sFlt-1-e15a, and reduced placental production and secretion of ADAM12 (a disintegrin and metalloproteinase12) and angiotensinogen. In vivo, galectin-7 administration (E8-E12) to pregnant mice caused elevated systolic blood pressure, albuminuria, impaired placentation (reduced labyrinth vascular branching, impaired decidual spiral artery remodeling, and a proinflammatory placental state demonstrated by elevated IL1β, IL6 and reduced IL10), and dysregulated expression of renin-angiotensin system components in the placenta, decidua, and kidney, including angiotensinogen, prorenin, and the angiotensin II type 1 receptor. Collectively, this study demonstrates that elevated galectin-7 during placental formation contributes to abnormal placentation and suggests that it leads to the development of preeclampsia via altering placental production of sFlt-1 and renin-angiotensin system components. Targeting galectin-7 may be a new treatment option for preeclampsia.
Collapse
Affiliation(s)
- Ellen Menkhorst
- From the Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia (E.M., W.Z., L.L.S., T.S., E.D.).,Gynaecology Research Centre, Royal Women's Hospital, Parkville, VIC, Australia (E.M., W.Z., L.L.S., T.S., E.D.).,Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia (E.M., K.R., H.L., E.D.)
| | - Wei Zhou
- From the Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia (E.M., W.Z., L.L.S., T.S., E.D.).,Gynaecology Research Centre, Royal Women's Hospital, Parkville, VIC, Australia (E.M., W.Z., L.L.S., T.S., E.D.)
| | - Leilani L Santos
- From the Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia (E.M., W.Z., L.L.S., T.S., E.D.).,Gynaecology Research Centre, Royal Women's Hospital, Parkville, VIC, Australia (E.M., W.Z., L.L.S., T.S., E.D.)
| | - Sarah Delforce
- School of Biomedical Sciences and Pharmacy (S.D., K.P.), University of Newcastle, NSW, Australia.,Priority Research Centre for Reproductive Sciences (S.D., K.P.), University of Newcastle, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, NSW, Australia (S.D., K.P.)
| | - Teresa So
- From the Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia (E.M., W.Z., L.L.S., T.S., E.D.).,Gynaecology Research Centre, Royal Women's Hospital, Parkville, VIC, Australia (E.M., W.Z., L.L.S., T.S., E.D.)
| | - Kate Rainczuk
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia (E.M., K.R., H.L., E.D.)
| | - Hannah Loke
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia (E.M., K.R., H.L., E.D.)
| | - Argyro Syngelaki
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom (A.S., K.H.N.)
| | - Swati Varshney
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, VIC, Australia (S.V., N.W.)
| | - Nicholas Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science & Biotechnology Institute, The University of Melbourne, VIC, Australia (S.V., N.W.)
| | - Kirsty Pringle
- School of Biomedical Sciences and Pharmacy (S.D., K.P.), University of Newcastle, NSW, Australia.,Priority Research Centre for Reproductive Sciences (S.D., K.P.), University of Newcastle, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, Newcastle, NSW, Australia (S.D., K.P.)
| | - Morag J Young
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, VIC, Australia (M.J.Y.).,Baker Heart & Diabetes Institute, Prahran, VIC, Australia (M.J.Y.)
| | - Kypros H Nicolaides
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom (A.S., K.H.N.)
| | - Yves St-Pierre
- INRS-Institut Armand-Frappier, Laval, QC, Canada (Y.S.-P.)
| | - Eva Dimitriadis
- From the Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville, VIC, Australia (E.M., W.Z., L.L.S., T.S., E.D.).,Gynaecology Research Centre, Royal Women's Hospital, Parkville, VIC, Australia (E.M., W.Z., L.L.S., T.S., E.D.).,Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia (E.M., K.R., H.L., E.D.).,Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia (E.D.)
| |
Collapse
|
30
|
Shi X, Xiao M, Xie Z, Shi Q, Zhang Y, Leavenworth JW, Yan B, Huang H. Angiostrongylus cantonensis Galectin-1 interacts with Annexin A2 to impair the viability of macrophages via activating JNK pathway. Parasit Vectors 2020; 13:183. [PMID: 32268913 PMCID: PMC7140382 DOI: 10.1186/s13071-020-04038-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/25/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Angiostrongylus cantonensis can cause severe symptoms of central nervous system infections. In the host, this parasite localizes in the blood and cerebrospinal fluid, and its secreted components can impact immune responses. Our previous study demonstrated that immune responses were inhibited in A. cantonensis-infected mice immunized with Ac-Galectin-1 (AcGal-1). However, the mechanisms by which AcGal-1 regulates the immune responses remain unclear. Macrophages are innate immune cells that rapidly respond to infection. The direct impact of AcGal-1 on macrophages may affect the immune responses. METHODS AcGal-1 protein was purified by nickel ion affinity chromatography. The effect of AcGal-1 on the apoptosis of macrophages was detected using CCK-8 assay, flow cytometry and western blot. Macrophage membrane proteins bound to AcGal-1 were obtained using the His-tag-based pull-down assay and identified via mass spectrometry. Co-localization of AcGal-1 and the macrophage membrane protein Annexin A2 was observed by immunofluorescence microscopy, and their interaction was validated by co-immunoprecipitation experiments. SiRNA-mediated knockdown of Annexin A2 was used to determine if AcGal-1-induced macrophage apoptosis required interaction with Annexin A2. The phosphorylation level of apoptotic signal pathway protein was detected by phospho-antibody microarray and western blot. RESULTS Our study showed that AcGal-1 caused apoptosis of the macrophages. AcGal-1 increased the expression of apoptosis proteins caspase-3, caspase-9, Bax, but reduced the expression of anti-apoptosis protein Bcl-2. AcGal-1 interacted with the membrane protein Annexin A2, and knockdown of Annexin A2 expression increased Bcl-2 but decreased Bax levels in AcGal-1-treated cells. Moreover, AcGal-1 increased JNK phosphorylation and the inhibition of JNK phosphorylation in AcGal-1-treated cells decreased the expression of caspase-3, -9, Bax and almost restored Bcl-2 to the level observed in control cells. CONCLUSIONS AcGal-1 can induce the apoptosis of macrophages by binding to Annexin A2 and activating JNK downstream the apoptotic signaling pathway.
Collapse
Affiliation(s)
- Xiaomeng Shi
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang People’s Republic of China
- The First Affiliated Hospital of Wenzhou Medical university, Wenzhou, 325035 Zhejiang People’s Republic of China
| | - Mengran Xiao
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang People’s Republic of China
| | - Zhiyue Xie
- The First Clinical College, Southern Medical University, Guangzhou, 510515 Guangdong People’s Republic of China
| | - Qing Shi
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang People’s Republic of China
| | - Yuanjiao Zhang
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang People’s Republic of China
| | - Jianmei W. Leavenworth
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Baolong Yan
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang People’s Republic of China
| | - Huicong Huang
- Department of Parasitology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang People’s Republic of China
| |
Collapse
|
31
|
Girotti MR, Salatino M, Dalotto-Moreno T, Rabinovich GA. Sweetening the hallmarks of cancer: Galectins as multifunctional mediators of tumor progression. J Exp Med 2020; 217:e20182041. [PMID: 31873723 DOI: 10.1084/jem_20182041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/14/2019] [Accepted: 11/18/2019] [Indexed: 01/03/2025] Open
Abstract
Hanahan and Weinberg have proposed 10 organizing principles that enable growth and metastatic dissemination of cancer cells. These distinctive and complementary capabilities, defined as the "hallmarks of cancer," include the ability of tumor cells and their microenvironment to sustain proliferative signaling, evade growth suppressors, resist cell death, promote replicative immortality, induce angiogenesis, support invasion and metastasis, reprogram energy metabolism, induce genomic instability and inflammation, and trigger evasion of immune responses. These common features are hierarchically regulated through different mechanisms, including those involving glycosylation-dependent programs that influence the biological and clinical impact of each hallmark. Galectins, an evolutionarily conserved family of glycan-binding proteins, have broad influence in tumor progression by rewiring intracellular and extracellular circuits either in cancer or stromal cells, including immune cells, endothelial cells, and fibroblasts. In this review, we dissect the role of galectins in shaping cellular circuitries governing each hallmark of tumors, illustrating relevant examples and highlighting novel opportunities for treating human cancer.
Collapse
Affiliation(s)
- María Romina Girotti
- Laboratorio de Inmuno-Oncología Traslacional, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Mariana Salatino
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Tomás Dalotto-Moreno
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
32
|
Saigusa R, Yamashita T, Miura S, Hirabayashi M, Nakamura K, Miyagawa T, Fukui Y, Yoshizaki A, Sato S, Asano Y. A potential contribution of decreased galectin‐7 expression in stratified epithelia to the development of cutaneous and oesophageal manifestations in systemic sclerosis. Exp Dermatol 2019; 28:536-542. [DOI: 10.1111/exd.13900] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 01/19/2019] [Accepted: 02/03/2019] [Indexed: 02/02/2023]
Affiliation(s)
- Ryosuke Saigusa
- Department of DermatologyUniversity of Tokyo Graduate School of Medicine Tokyo Japan
| | - Takashi Yamashita
- Department of DermatologyUniversity of Tokyo Graduate School of Medicine Tokyo Japan
| | - Shunsuke Miura
- Department of DermatologyUniversity of Tokyo Graduate School of Medicine Tokyo Japan
| | - Megumi Hirabayashi
- Department of DermatologyUniversity of Tokyo Graduate School of Medicine Tokyo Japan
| | - Kouki Nakamura
- Department of DermatologyUniversity of Tokyo Graduate School of Medicine Tokyo Japan
| | - Takuya Miyagawa
- Department of DermatologyUniversity of Tokyo Graduate School of Medicine Tokyo Japan
| | - Yuki Fukui
- Department of DermatologyUniversity of Tokyo Graduate School of Medicine Tokyo Japan
| | - Ayumi Yoshizaki
- Department of DermatologyUniversity of Tokyo Graduate School of Medicine Tokyo Japan
| | - Shinichi Sato
- Department of DermatologyUniversity of Tokyo Graduate School of Medicine Tokyo Japan
| | - Yoshihide Asano
- Department of DermatologyUniversity of Tokyo Graduate School of Medicine Tokyo Japan
| |
Collapse
|
33
|
Asano Y, Takahashi T, Saigusa R. Systemic sclerosis: Is the epithelium a missing piece of the pathogenic puzzle? J Dermatol Sci 2019; 94:259-265. [DOI: 10.1016/j.jdermsci.2019.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 04/26/2019] [Indexed: 10/26/2022]
|
34
|
Wu NL, Liu FT. The expression and function of galectins in skin physiology and pathology. Exp Dermatol 2019; 27:217-226. [PMID: 29427464 DOI: 10.1111/exd.13512] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2018] [Indexed: 01/01/2023]
Abstract
The galectin family comprises β-galactoside-binding proteins widely expressed in many organisms. There are at least 16 family members, which can be classified into three groups based on their carbohydrate-recognition domains. Pleiotropic functions of different galectins in physiological and pathological processes through extracellular or intracellular actions have been revealed. In the skin, galectins are expressed in a variety of cells, including keratinocytes, melanocytes, fibroblasts, dendritic cells, lymphocytes, macrophages and endothelial cells. Expression of specific galectins is reported to affect cell status, such as activation or death, and regulate the interaction between different cell types or between cells and the extracellular matrix. In vitro cellular studies, in vivo animal studies and studies of human clinical material have revealed the pathophysiologic roles of galectins in the skin. The pathogenesis of diverse non-malignant skin disorders, such as atopic dermatitis, psoriasis, contact dermatitis and wound healing, as well as skin cancers, such as melanoma, squamous cell carcinoma, basal cell carcinoma and cutaneous haematologic malignancy can be regulated by different galectins. Revelation of biological roles of galectins in skin may pave the way to future development of galectin-based therapeutic strategies for skin diseases.
Collapse
Affiliation(s)
- Nan-Lin Wu
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan.,Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan.,Mackay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Department of Dermatology, University of California Davis, Sacramento, CA, USA
| |
Collapse
|
35
|
Tian Y, Li H, Gao Y, Liu C, Qiu T, Wu H, Cao M, Zhang Y, Ding H, Chen J, Cai H. Quantitative proteomic characterization of lung tissue in idiopathic pulmonary fibrosis. Clin Proteomics 2019; 16:6. [PMID: 30774578 PMCID: PMC6364390 DOI: 10.1186/s12014-019-9226-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/27/2019] [Indexed: 02/06/2023] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a progressive, eventually fatal disease. IPF is characterized by excessive accumulation of the extracellular matrix (ECM) in the alveolar parenchyma and progressive lung scarring. The pathogenesis of IPF and whether the ECM involved in the process remain unknown. Methods To identify potential treatment target and ECM associated proteins that may be involved in the development of IPF, we employed isobaric tag for relative and absolute quantitation (iTRAQ) combined liquid chromatography-tandem mass spectrometry (LC-MS/MS) approach to examine protein expression in lung tissues from IPF patients. Results A total of 662 proteins with altered expression (455 upregulated proteins and 207 downregulated proteins) were identified in lung tissue of IPF patients compared with control. KEGG pathway enrichment analysis showed that the altered proteins in lung tissue mainly belonged to the PI3K-Akt signaling, focal adhesion, ECM-receptor interaction, and carbon metabolism pathways. According to the bioinformatic definition of the matrisome, 229 matrisome proteins were identified in lung tissue. These proteins comprised the ECM of lung, of which 104 were core matrisome proteins, and 125 were matrisome-associated proteins. Of the 229 ECM quantified proteins, 56 significantly differentially expressed proteins (19 upregulated proteins and 37 downregulated proteins) were detected in IPF lung tissue samples. In addition to proteins with well-known functions such as COL1A1, SCGB1A1, TAGLN, PSEN2, TSPAN1, CTSB, AGR2, CSPG2, and SERPINB3, we identified several novel ECM proteins with unknown function deposited in IPF lung tissue including LGALS7, ASPN, HSP90AA1 and HSP90AB1. Some of these differentially expressed proteins were further verified using Western blot analysis and immunohistochemical staining. Conclusions This study provides a list of proteomes that were detected in IPF lung tissue by iTRAQ technology combined with LC-MS/MS. The findings of this study will contribute better understanding to the pathogenesis of IPF and facilitate the development of therapeutic targets.
Collapse
Affiliation(s)
- Yaqiong Tian
- 1Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008 Jiangsu People's Republic of China
| | - Hui Li
- 1Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008 Jiangsu People's Republic of China
| | - Yujuan Gao
- 1Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008 Jiangsu People's Republic of China
| | - Chuanmei Liu
- 2Department of Respiratory Medicine, Yi Ji Shan Hospital of Wannan Medical College, No. 2 Zheshan West Road, Wuhu, 241001 Anhui People's Republic of China
| | - Ting Qiu
- Department of Respiratory Medicine, KunShan Hospital of Traditional Chinese Medicine, No. 189 Chaoyang Road, Kunshan, 215300 Jiangsu People's Republic of China
| | - Hongyan Wu
- 4Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008 Jiangsu People's Republic of China
| | - Mengshu Cao
- 1Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008 Jiangsu People's Republic of China
| | - Yingwei Zhang
- 1Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008 Jiangsu People's Republic of China
| | - Hui Ding
- 5Department of Respiratory Medicine, Yixing People Hospital, Affiliated Jiangsu University, No. 75 Tongzhenguan Road, Yixing, 214200 Jiangsu People's Republic of China
| | - Jingyu Chen
- 6Jiangsu Key Laboratory of Organ Transplantation, Wuxi People's Hospital, Nanjing Medical University, No. 299 Qingyang Road, Wuxi, 214023 Jiangsu People's Republic of China
| | - Hourong Cai
- 1Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008 Jiangsu People's Republic of China
| |
Collapse
|
36
|
The Role of Galectins as Modulators of Metabolism and Inflammation. Mediators Inflamm 2018; 2018:9186940. [PMID: 29950926 PMCID: PMC5987346 DOI: 10.1155/2018/9186940] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/19/2018] [Accepted: 05/09/2018] [Indexed: 12/15/2022] Open
Abstract
Galectins are β-galcotosid-binding lectins. The function of galectins varies with their tissue-specific and subcellular location, and their binding to carbohydrates makes them key players in several intra- and extracellular processes where they bind to glycosylated proteins and lipids. In humans, there are 12 identified galectins, some with tissue-specific distribution. Galectins are found inside cells and in the nucleus, cytosol, and organelles, as well as extracellularly. Galectin-1, -2, -3, -4, -7, -8, -9, and -12 can all induce T-cell apoptosis and modulate inflammation. In the context of metabolic control and loss of the same in, for example, diabetes, galectin-1, -2, -3, -9, and -12 are especially interesting. This review presents information on galectins relevant to the control of inflammation and metabolism and the potential to target galectins for therapeutic purposes.
Collapse
|
37
|
Choi JW, Nam KM, Choi HR, Lee DH, Huh CH, Park KC. Decreased Galectin-3 and -7 Expressions in Old-Aged Skin and Their Differential Expression in Skin Equivalents. Ann Dermatol 2018; 30:375-378. [PMID: 29853761 PMCID: PMC5929964 DOI: 10.5021/ad.2018.30.3.375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/24/2017] [Accepted: 06/11/2017] [Indexed: 12/02/2022] Open
Affiliation(s)
- Jee Woong Choi
- Department of Dermatology, Ajou University School of Medicine, Suwon, Korea
| | - Kyung Mi Nam
- Department of Dermatology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hye Ryung Choi
- Department of Dermatology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Chang Hun Huh
- Department of Dermatology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Kyung Chan Park
- Department of Dermatology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
38
|
Sundblad V, Quintar AA, Morosi LG, Niveloni SI, Cabanne A, Smecuol E, Mauriño E, Mariño KV, Bai JC, Maldonado CA, Rabinovich GA. Galectins in Intestinal Inflammation: Galectin-1 Expression Delineates Response to Treatment in Celiac Disease Patients. Front Immunol 2018; 9:379. [PMID: 29545799 PMCID: PMC5837985 DOI: 10.3389/fimmu.2018.00379] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 02/12/2018] [Indexed: 12/21/2022] Open
Abstract
Galectins, a family of animal lectins characterized by their affinity for N-acetyllactosamine-enriched glycoconjugates, modulate several immune cell processes shaping the course of innate and adaptive immune responses. Through interaction with a wide range of glycosylated receptors bearing complex branched N-glycans and core 2-O-glycans, these endogenous lectins trigger distinct signaling programs thereby controling immune cell activation, differentiation, recruitment and survival. Given the unique features of mucosal inflammation and the differential expression of galectins throughout the gastrointestinal tract, we discuss here key findings on the role of galectins in intestinal inflammation, particularly Crohn’s disease, ulcerative colitis, and celiac disease (CeD) patients, as well as in murine models resembling these inflammatory conditions. In addition, we present new data highlighting the regulated expression of galectin-1 (Gal-1), a proto-type member of the galectin family, during intestinal inflammation in untreated and treated CeD patients. Our results unveil a substantial upregulation of Gal-1 accompanying the anti-inflammatory and tolerogenic response associated with gluten-free diet in CeD patients, suggesting a major role of this lectin in favoring resolution of inflammation and restoration of mucosal homeostasis. Thus, a coordinated network of galectins and their glycosylated ligands, exerting either anti-inflammatory or proinflammatory responses, may influence the interplay between intestinal epithelial cells and the highly specialized gut immune system in physiologic and pathologic settings.
Collapse
Affiliation(s)
- Victoria Sundblad
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Amado A Quintar
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Luciano G Morosi
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental (IBYME), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Sonia I Niveloni
- Sección Intestino Delgado, Departamento de Medicina, Hospital de Gastroenterología Dr. C. Bonorino Udaondo, Buenos Aires, Argentina
| | - Ana Cabanne
- Unidad de Patología, Hospital de Gastroenterología, Bonorino Udaondo, Buenos Aires, Argentina
| | - Edgardo Smecuol
- Sección Intestino Delgado, Departamento de Medicina, Hospital de Gastroenterología Dr. C. Bonorino Udaondo, Buenos Aires, Argentina
| | - Eduardo Mauriño
- Sección Intestino Delgado, Departamento de Medicina, Hospital de Gastroenterología Dr. C. Bonorino Udaondo, Buenos Aires, Argentina
| | - Karina V Mariño
- Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental (IBYME), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Julio C Bai
- Sección Intestino Delgado, Departamento de Medicina, Hospital de Gastroenterología Dr. C. Bonorino Udaondo, Buenos Aires, Argentina.,Instituto de Investigaciones, Universidad del Salvador, Buenos Aires, Argentina
| | - Cristina A Maldonado
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
39
|
Watanabe K, Yoshino T, Takahashi M, Kondo S, Takahashi M, Matsukuma S, Mukai H, Fukuda H. Clinical significance of neutrophil gelatinase-associated lipocalin and galectin-7 in tape-stripped stratum corneum of acne vulgaris. J Dermatol 2018; 45:618-621. [PMID: 29473202 DOI: 10.1111/1346-8138.14261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/18/2018] [Indexed: 11/26/2022]
Abstract
The usefulness of stratum corneum neutrophil gelatinase-associated lipocalin and stratum corneum galectin-7 as biomarkers of acne vulgaris was studied. A comparison of neutrophil gelatinase-associated lipocalin levels on the cheeks of patients with acne vulgaris at the start of the study and at the time of symptom improvement showed a significant decrease. On the other hand, the galectin-7 levels at the time of symptom improvement were significantly higher than those at the start of the study. Therefore, because the inflammation in the epidermis and hair follicles was reduced after therapy, as a result of the solution of the inflammatory eruptions caused by acne vulgaris, the neutrophil gelatinase-associated lipocalin level also showed a significant decrease after therapy. These results suggest that stratum corneum neutrophil gelatinase-associated lipocalin may be useful as an objective biomarker of changes in acne vulgaris symptoms.
Collapse
Affiliation(s)
- Kosuke Watanabe
- Department of Dermatology, Toho University Ohashi Medical Center, Tokyo, Japan
| | | | - Misaki Takahashi
- Department of Dermatology, Toho University Ohashi Medical Center, Tokyo, Japan
| | | | | | | | - Hideki Mukai
- Department of Dermatology, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Hidetsugu Fukuda
- Department of Dermatology, Toho University Ohashi Medical Center, Tokyo, Japan
| |
Collapse
|
40
|
Dissecting the Structure-Activity Relationship of Galectin-Ligand Interactions. Int J Mol Sci 2018; 19:ijms19020392. [PMID: 29382172 PMCID: PMC5855614 DOI: 10.3390/ijms19020392] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 01/18/2018] [Accepted: 01/24/2018] [Indexed: 02/08/2023] Open
Abstract
Galectins are β-galactoside-binding proteins. As carbohydrate-binding proteins, they participate in intracellular trafficking, cell adhesion, and cell-cell signaling. Accumulating evidence indicates that they play a pivotal role in numerous physiological and pathological activities, such as the regulation on cancer progression, inflammation, immune response, and bacterial and viral infections. Galectins have drawn much attention as targets for therapeutic interventions. Several molecules have been developed as galectin inhibitors. In particular, TD139, a thiodigalactoside derivative, is currently examined in clinical trials for the treatment of idiopathic pulmonary fibrosis. Herein, we provide an in-depth review on the development of galectin inhibitors, aiming at the dissection of the structure-activity relationship to demonstrate how inhibitors interact with galectin(s). We especially integrate the structural information established by X-ray crystallography with several biophysical methods to offer, not only in-depth understanding at the molecular level, but also insights to tackle the existing challenges.
Collapse
|
41
|
Galectin-7 in Epithelial Homeostasis and Carcinomas. Int J Mol Sci 2017; 18:ijms18122760. [PMID: 29257082 PMCID: PMC5751359 DOI: 10.3390/ijms18122760] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 01/23/2023] Open
Abstract
Galectins are small unglycosylated soluble lectins distributed both inside and outside the cells. They share a conserved domain for the recognition of carbohydrates (CRD). Although galectins have a common affinity for β-galatosides, they exhibit different binding preferences for complex glycans. First described twenty years ago, galectin-7 is a prototypic galectin, with a single CRD, able to form divalent homodimers. This lectin, which is mainly expressed in stratified epithelia, has been described in epithelial tissues as being involved in apoptotic responses, in proliferation and differentiation but also in cell adhesion and migration. Most members of the galectins family have been associated with cancer biology. One of the main functions of galectins in cancer is their immunomodulating potential and anti-angiogenic activity. Indeed, galectin-1 and -3, are already targeted in clinical trials. Another relevant function of galectins in tumour progression is their ability to regulate cell migration and cell adhesion. Among these galectins, galectin-7 is abnormally expressed in various cancers, most prominently in carcinomas, and is involved in cancer progression and metastasis but its precise functions in tumour biology remain poorly understood. In this issue, we will focus on the physiological functions of galectin-7 in epithelia and present the alterations of galectin-7 expression in carcinomas with the aim to describe its possible functions in tumour progression.
Collapse
|
42
|
Advedissian T, Proux-Gillardeaux V, Nkosi R, Peyret G, Nguyen T, Poirier F, Viguier M, Deshayes F. E-cadherin dynamics is regulated by galectin-7 at epithelial cell surface. Sci Rep 2017; 7:17086. [PMID: 29213102 PMCID: PMC5719072 DOI: 10.1038/s41598-017-17332-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/15/2017] [Indexed: 02/02/2023] Open
Abstract
Re-epithelialisation of wounded epidermis is ensured by collective cell migration of keratinocytes. Efficient collective migration requires the maintenance of intercellular adhesion, notably through adherens junctions, to favour cell communication, support tension forces and coordinated movement . Galectin-7, a soluble lectin expressed in stratified epithelia, has been previously implicated in cell migration and intercellular adhesion. Here, we revealed a new function of galectin-7 in the control of directionality and collective behaviour in migrating keratinocytes. Consistently, we identified galectin-7 as a direct partner of E-cadherin, a key component of adherens junctions. Unexpectedly, this interaction does not require glycosylation motifs. Focusing on the underlying mechanisms, we showed that galectin-7 stabilizes E-cadherin at the plasma membrane, restraining its endocytosis. Interestingly, galectin-7 silencing decreases E-cadherin-mediated intercellular adhesion. Consequently, this study not only identifies a new stabilizer of adherens junctions but also emphasises the importance of the interplay between E-cadherin turnover and intercellular adhesion strength.
Collapse
Affiliation(s)
- Tamara Advedissian
- Team Morphogenesis, Homeostasis and Pathologies, University Paris Diderot, Sorbonne Paris Cité, CNRS UMR 7592, Institut Jacques Monod, 15 Rue Hélène Brion, 75013, Paris, France
| | - Véronique Proux-Gillardeaux
- Team Morphogenesis, Homeostasis and Pathologies, University Paris Diderot, Sorbonne Paris Cité, CNRS UMR 7592, Institut Jacques Monod, 15 Rue Hélène Brion, 75013, Paris, France.,Team Membrane Traffic in Health & Disease, University Paris Diderot, Sorbonne Paris Cité, CNRS UMR 7592, Institut Jacques Monod, 15 Rue Hélène Brion, 75013, Paris, France
| | - Rachel Nkosi
- Team Morphogenesis, Homeostasis and Pathologies, University Paris Diderot, Sorbonne Paris Cité, CNRS UMR 7592, Institut Jacques Monod, 15 Rue Hélène Brion, 75013, Paris, France
| | - Grégoire Peyret
- Team Cell Adhesion and Mechanics, University Paris Diderot, Sorbonne Paris Cité, CNRS UMR 7592, Institut Jacques Monod, 15 Rue Hélène Brion, 75013, Paris, France
| | - Thao Nguyen
- Team Cell Adhesion and Mechanics, University Paris Diderot, Sorbonne Paris Cité, CNRS UMR 7592, Institut Jacques Monod, 15 Rue Hélène Brion, 75013, Paris, France
| | - Françoise Poirier
- Team Morphogenesis, Homeostasis and Pathologies, University Paris Diderot, Sorbonne Paris Cité, CNRS UMR 7592, Institut Jacques Monod, 15 Rue Hélène Brion, 75013, Paris, France
| | - Mireille Viguier
- Team Morphogenesis, Homeostasis and Pathologies, University Paris Diderot, Sorbonne Paris Cité, CNRS UMR 7592, Institut Jacques Monod, 15 Rue Hélène Brion, 75013, Paris, France.
| | - Frédérique Deshayes
- Team Morphogenesis, Homeostasis and Pathologies, University Paris Diderot, Sorbonne Paris Cité, CNRS UMR 7592, Institut Jacques Monod, 15 Rue Hélène Brion, 75013, Paris, France.
| |
Collapse
|
43
|
Dion J, Advedissian T, Storozhylova N, Dahbi S, Lambert A, Deshayes F, Viguier M, Tellier C, Poirier F, Téletchéa S, Dussouy C, Tateno H, Hirabayashi J, Grandjean C. Development of a Sensitive Microarray Platform for the Ranking of Galectin Inhibitors: Identification of a Selective Galectin-3 Inhibitor. Chembiochem 2017; 18:2428-2440. [DOI: 10.1002/cbic.201700544] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Johann Dion
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2, chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Tamara Advedissian
- Université Paris Diderot-Paris 7 Sorbonne Paris Cité; Institut Jacques Monod; UMR CNRS 7592; 15 rue Hélène Brion 75205 Paris Cedex 13 France
| | - Nataliya Storozhylova
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2, chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Samir Dahbi
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2, chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Annie Lambert
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2, chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Frédérique Deshayes
- Université Paris Diderot-Paris 7 Sorbonne Paris Cité; Institut Jacques Monod; UMR CNRS 7592; 15 rue Hélène Brion 75205 Paris Cedex 13 France
| | - Mireille Viguier
- Université Paris Diderot-Paris 7 Sorbonne Paris Cité; Institut Jacques Monod; UMR CNRS 7592; 15 rue Hélène Brion 75205 Paris Cedex 13 France
| | - Charles Tellier
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2, chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Françoise Poirier
- Université Paris Diderot-Paris 7 Sorbonne Paris Cité; Institut Jacques Monod; UMR CNRS 7592; 15 rue Hélène Brion 75205 Paris Cedex 13 France
| | - Stéphane Téletchéa
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2, chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Christophe Dussouy
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2, chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Hiroaki Tateno
- Biotechnology Research Institute for Drug Discovery; Cellular Glycome-Targeted Technology Group; National Institute of Advanced Industrial Science and Technology (AIST); Central 2 1-1-1 Umezuno Tsukuba Ibaraki 305-8568 Japan
| | - Jun Hirabayashi
- Biotechnology Research Institute for Drug Discovery; Cellular Glycome-Targeted Technology Group; National Institute of Advanced Industrial Science and Technology (AIST); Central 2 1-1-1 Umezuno Tsukuba Ibaraki 305-8568 Japan
| | - Cyrille Grandjean
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2, chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| |
Collapse
|
44
|
Itoh A, Nonaka Y, Ogawa T, Nakamura T, Nishi N. Small leucine-rich repeat proteoglycans associated with mature insoluble elastin serve as binding sites for galectins. Biosci Biotechnol Biochem 2017; 81:2098-2104. [DOI: 10.1080/09168451.2017.1374828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Abstract
We previously reported that galectin-9 (Gal-9), an immunomodulatory animal lectin, could bind to insoluble collagen preparations and exerted direct cytocidal effects on immune cells. In the present study, we found that mature insoluble elastin is capable of binding Gal-9 and other members of the human galectin family. Lectin blot analysis of a series of commercial water-soluble elastin preparations, PES-(A) ~ PES-(E), revealed that only PES-(E) contained substances recognized by Gal-9. Gal-9-interacting substances in PES-(E) were affinity-purified, digested with trypsin and then analyzed by reversed-phase HPLC. Peptide fragments derived from five members of the small leucine-rich repeat proteoglycan family, versican, lumican, osteoglycin/mimecan, prolargin, and fibromodulin, were identified by N-terminal amino acid sequence analysis. The results indicate that Gal-9 and possibly other galectins recognize glycans attached to small leucine-rich repeat proteoglycans associated with insoluble elastin and also indicate the possibility that mature insoluble elastin serves as an extracellular reservoir for galectins.
Collapse
Affiliation(s)
- Aiko Itoh
- Division of Research Instrument and Equipment, Life Science Research Center, Kagawa University, Kagawa, Japan
| | - Yasuhiro Nonaka
- Department of Endocrinology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takashi Ogawa
- Department of Endocrinology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takanori Nakamura
- Department of Endocrinology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Nozomu Nishi
- Division of Research Instrument and Equipment, Life Science Research Center, Kagawa University, Kagawa, Japan
| |
Collapse
|
45
|
Huang SM, Wu CS, Chiu MH, Yang HJ, Chen GS, Lan CCE. High-glucose environment induced intracellular O-GlcNAc glycosylation and reduced galectin-7 expression in keratinocytes: Implications on impaired diabetic wound healing. J Dermatol Sci 2017; 87:168-175. [PMID: 28526214 DOI: 10.1016/j.jdermsci.2017.04.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/16/2017] [Accepted: 04/26/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Diabetes is an important global health issue due to its increasing prevalence and association with various complications. Impaired wound healing is a serious complication associated with diabetes that frequently results in infection and amputation. Galectin-7 (Gal-7) has been reported to play an important role during skin wound healing. Previously, we had demonstrated that high glucose environment alters physiologic functions of keratinocytes and contributes to impaired wound healing in diabetic condition. OBJECTIVE In this study, we hypothesized that Gal-7 expression of keratinocytes may be involved in delayed wound healing of diabetics. METHODS Using cultured human keratinocytes and diabetic mice model, the Gal-7 expression was evaluated under high glucose environment. RESULTS Our results demonstrated that high-glucose environment reduced Gal-7 expression, a molecule that plays an important role in keratinocyte migration. Additionally, we found that increased O-linked N-Acetyl-glucosamine (O-GlcNAc) is responsible for reduced Gal-7 expression in keratinocytes exposed to high glucose environment. CONCLUSION Taken together, restoring the levels of Gal-7 and O-GlcNAc glycosylation may present novel therapeutic approach to promote wound healing in diabetic patients.
Collapse
Affiliation(s)
- Shu-Mei Huang
- Department of Dermatology, Kaohsiung Medical University Hospital, Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Shuang Wu
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Min-Hsi Chiu
- Department of Dermatology, Kaohsiung Medical University Hospital, Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Jun Yang
- Department of Dermatology, Kaohsiung Medical University Hospital, Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Gwo-Shing Chen
- Department of Dermatology, Kaohsiung Medical University Hospital, Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng-Che E Lan
- Department of Dermatology, Kaohsiung Medical University Hospital, Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
46
|
Dion J, Deshayes F, Storozhylova N, Advedissian T, Lambert A, Viguier M, Tellier C, Dussouy C, Poirier F, Grandjean C. Lactosamine-Based Derivatives as Tools to Delineate the Biological Functions of Galectins: Application to Skin Tissue Repair. Chembiochem 2017; 18:782-789. [DOI: 10.1002/cbic.201600673] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Indexed: 11/08/2022]
Affiliation(s)
- Johann Dion
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2 chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Frédérique Deshayes
- Université Paris Diderot-Paris 7 Sorbonne Paris Cité; Institut Jacques Monod; UMR CNRS 7592; 15 rue Hélène Brion 75205 Paris Cedex 13 France
| | - Nataliya Storozhylova
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2 chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Tamara Advedissian
- Université Paris Diderot-Paris 7 Sorbonne Paris Cité; Institut Jacques Monod; UMR CNRS 7592; 15 rue Hélène Brion 75205 Paris Cedex 13 France
| | - Annie Lambert
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2 chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Mireille Viguier
- Université Paris Diderot-Paris 7 Sorbonne Paris Cité; Institut Jacques Monod; UMR CNRS 7592; 15 rue Hélène Brion 75205 Paris Cedex 13 France
| | - Charles Tellier
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2 chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Christophe Dussouy
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2 chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| | - Françoise Poirier
- Université Paris Diderot-Paris 7 Sorbonne Paris Cité; Institut Jacques Monod; UMR CNRS 7592; 15 rue Hélène Brion 75205 Paris Cedex 13 France
| | - Cyrille Grandjean
- Faculté des Sciences et des Techniques; Unité Fonctionnalité et Ingénierie des Protéines (UFIP); Université de Nantes; UMR CNRS 6286; 2 chemin de la Houssinière B. P. 92208 44322 Nantes Cedex 3 France
| |
Collapse
|
47
|
Grosset AA, Poirier F, Gaboury L, St-Pierre Y. Galectin-7 Expression Potentiates HER-2-Positive Phenotype in Breast Cancer. PLoS One 2016; 11:e0166731. [PMID: 27902734 PMCID: PMC5130216 DOI: 10.1371/journal.pone.0166731] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/31/2016] [Indexed: 01/03/2023] Open
Abstract
HER-2 positive tumors are among the most aggressive subtypes of breast cancer and are frequently associated with metastasis and poor outcome. As with other aggressive subtypes of breast cancer, these tumors are associated with abnormally high expression of galectin-7 (gal-7), which confers metastatic breast tumor cells with increased invasive behavior. Although previous studies in the rat model of breast tumorigenesis have shown that gal-7 is also increased in primary breast tumor, its contribution to the development of the primary breast tumors remains unclear. In the present work, we have used genetically-engineered gal-7-deficient mice to examine the role of gal-7 in the development of the mammary gland and of breast cancer. Using histological and immunohistological analysis of whole mammary glands at different stages of development, we detected no significant changes between normal and gal-7-deficient mice. To test the involvement of gal-7 in breast cancer, we next examined the effects of loss of gal-7 on mammary tumor development by crossing gal-7-deficient mice with the mammary tumor transgenic mouse strain FVB-Tg(MMTV-Erbb2)NK1Mul/J. Finally, assessment of mice survival and tumor volume showed a delay of mammary tumor growth in the absence of systemic gal-7. These data suggest that gal-7 could potentiate the phenotype of HER-2 positive primary breast cancer.
Collapse
Affiliation(s)
- Andrée-Anne Grosset
- INRS-Institut Armand-Frappier, Laval, QC, Canada.,IRIC
- Université de Montréal, Montreal, QC, Canada
| | - Françoise Poirier
- Institut Jacques Monod, CNRS, Univ Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | | |
Collapse
|
48
|
Systems-level effects of ectopic galectin-7 reconstitution in cervical cancer and its microenvironment. BMC Cancer 2016; 16:680. [PMID: 27558259 PMCID: PMC4997669 DOI: 10.1186/s12885-016-2700-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022] Open
Abstract
Background Galectin-7 (Gal-7) is negatively regulated in cervical cancer, and appears to be a link between the apoptotic response triggered by cancer and the anti-tumoral activity of the immune system. Our understanding of how cervical cancer cells and their molecular networks adapt in response to the expression of Gal-7 remains limited. Methods Meta-analysis of Gal-7 expression was conducted in three cervical cancer cohort studies and TCGA. In silico prediction and bisulfite sequencing were performed to inquire epigenetic alterations. To study the effect of Gal-7 on cervical cancer, we ectopically re-expressed it in the HeLa and SiHa cervical cancer cell lines, and analyzed their transcriptome and SILAC-based proteome. We also examined the tumor and microenvironment host cell transcriptomes after xenotransplantation into immunocompromised mice. Differences between samples were assessed with the Kruskall-Wallis, Dunn’s Multiple Comparison and T tests. Kaplan–Meier and log-rank tests were used to determine overall survival. Results Gal-7 was constantly downregulated in our meta-analysis (p < 0.0001). Tumors with combined high Gal-7 and low galectin-1 expression (p = 0.0001) presented significantly better prognoses (p = 0.005). In silico and bisulfite sequencing assays showed de novo methylation in the Gal-7 promoter and first intron. Cells re-expressing Gal-7 showed a high apoptosis ratio (p < 0.05) and their xenografts displayed strong growth retardation (p < 0.001). Multiple gene modules and transcriptional regulators were modulated in response to Gal-7 reconstitution, both in cervical cancer cells and their microenvironments (FDR < 0.05 %). Most of these genes and modules were associated with tissue morphogenesis, metabolism, transport, chemokine activity, and immune response. These functional modules could exert the same effects in vitro and in vivo, even despite different compositions between HeLa and SiHa samples. Conclusions Gal-7 re-expression affects the regulation of molecular networks in cervical cancer that are involved in diverse cancer hallmarks, such as metabolism, growth control, invasion and evasion of apoptosis. The effect of Gal-7 extends to the microenvironment, where networks involved in its configuration and in immune surveillance are particularly affected. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2700-8) contains supplementary material, which is available to authorized users.
Collapse
|
49
|
Niiyama S, Yoshino T, Yasuda C, Yu X, Izumi R, Ishiwatari S, Matsukuma S, Mukai H. Galectin-7 in the stratum corneum: a biomarker of the skin barrier function. Int J Cosmet Sci 2016; 38:487-95. [PMID: 27028525 DOI: 10.1111/ics.12326] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 03/19/2016] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Skin barrier disruption often occurs in diseased and damaged skin conditions such as atopic dermatitis (AD). We focused the galectin-7 protein (Gal-7) as a biomarker of skin condition and assessed whether the content of Gal-7 in stratum corneum (scGal-7) could be used as an indicator of skin barrier disruption and as an index of local skin symptoms in AD patients. METHODS Alteration of Gal-7 expression levels in keratinocyte and scGal-7 contents after barrier disruption by sodium dodecyl sulphate were evaluated in vitro and in vivo, respectively. Correlation between scGal-7 content and transepidermal water loss (TEWL) was examined in 126 healthy subjects. We performed single measurements of scGal-7 contents in 34 AD patients and serial measurements of 15 inpatients among them. SC samples were collected by the tape-stripping method, and scGal-7 content was determined using enzyme-linked immunosorbent assay. RESULTS Gal-7 expression in keratinocytes increased after barrier disruption. The scGal-7 content reflected the disruption of the skin barrier. The scGal-7 contents and TEWL values correlated in healthy subjects. The scGal-7 level was higher in AD patients than in healthy subjects. The scGal-7 contents in the cheek and neck of AD patients significantly correlated with the total and local skin lesion severity scores. Serial measurements in the inpatients showed that the scGal-7 contents in the cheek and neck decreased in tandem with local severity scores in response to treatment. CONCLUSION Measurement of scGal-7 content in tape-stripped samples was useful for the evaluation of the skin barrier function in dry skin conditions such as AD.
Collapse
Affiliation(s)
- S Niiyama
- Department of Dermatology, Toho University Ohashi Medical Center, Tokyo, Japan.
| | - T Yoshino
- Fancl Research Institute, Yokohama, Japan
| | - C Yasuda
- Fancl Research Institute, Yokohama, Japan
| | - X Yu
- Fancl Research Institute, Yokohama, Japan
| | - R Izumi
- Fancl Research Institute, Yokohama, Japan
| | | | | | - H Mukai
- Department of Dermatology, Toho University Ohashi Medical Center, Tokyo, Japan
| |
Collapse
|
50
|
Chen HL, Chiang PC, Lo CH, Lo YH, Hsu DK, Chen HY, Liu FT. Galectin-7 Regulates Keratinocyte Proliferation and Differentiation through JNK-miR-203-p63 Signaling. J Invest Dermatol 2016; 136:182-191. [PMID: 26763438 PMCID: PMC4803640 DOI: 10.1038/jid.2015.366] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 08/19/2015] [Accepted: 08/22/2015] [Indexed: 02/07/2023]
Abstract
Galectin-7, a member of the β-galactoside-binding protein family, is primarily expressed in stratified epithelial cells, including keratinocytes. There is information in the literature suggesting a role for this protein in regulation of keratinocyte survival and growth, but the underlying mechanism remains relatively unknown. Moreover, its expression pattern in the epidermis suggests that it is also involved in the regulation of keratinocyte differentiation. Here, we demonstrate that galectin-7 knockdown results in reduced differentiation and increased proliferation of keratinocytes. Using microarray and deep-sequencing analyses, we found that galectin-7 positively and negatively regulates microRNA (miR)-203 and miR-146a expression, respectively. We show that galectin-7 regulates keratinocyte differentiation and proliferation through miR-203 but not miR-146a. A knockdown of either galectin-7 or miR-203 in keratinocytes increases expression of p63, an essential transcription factor involved in skin development. Rescue of miR-203 expression in a galectin-7 knockdown model reduces p63 expression to baseline. Increased galectin-7 expression upregulates c-Jun N-terminal kinase (JNK) protein levels, which is required for miR-203 expression. Finally, we establish that galectin-7 can be associated with JNK1 and protect it from ubiquitination and degradation. Thus, our data suggest an intracellular function of galectin-7: regulation of keratinocyte proliferation and differentiation through the JNK1-miR-203-p63 pathway.
Collapse
Affiliation(s)
- Hung-Lin Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Po-Cheng Chiang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Chia-Hui Lo
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yuan-Hsin Lo
- Graduate institute of immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Daniel K Hsu
- Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, USA
| | - Huan-Yuan Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Fu-Tong Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Graduate institute of immunology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Dermatology, School of Medicine, University of California-Davis, Sacramento, California, USA.
| |
Collapse
|