1
|
Hasan ML, Lee JR, Rahaman KA, Yang DH, Joung YK. Versatile effects of galectin-1 protein-containing lipid bilayer coating for cardiovascular applications. Bioact Mater 2024; 42:207-225. [PMID: 39285911 PMCID: PMC11403261 DOI: 10.1016/j.bioactmat.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024] Open
Abstract
Modulating inflammatory cells in an implantation site leads to severe complications and still unsolved challenges for blood-contacting medical devices. Inspired by the role of galectin-1 (Gal-1) in selective functions on multiple cells and immunomodulatory processes, we prepared a biologically target-specific surface coated with the lipid bilayer containing Gal-1 (Gal-1-SLB) and investigate the proof of the biological effects. First, lipoamido-dPEG-acid was deposited on a gold-coated substrate to form a self-assembled monolayer and then conjugated dioleoylphosphatidylethanolamine (DOPE) onto that to produce a lower leaflet of the supported lipid bilayer (SLB) before fusing membrane-derived vesicles extracted from B16-F10 cells. The Gal-1-SLB showed the expected anti-fouling activity by revealing the resistance to protein adsorption and bacterial adhesion. In vitro studies showed that the Gal-1-SLB can promote endothelial function and inhibit smooth muscle cell proliferation. Moreover, Gal-1- SLB presents potential function for endothelial cell migration and angiogenic activities. In vitro macrophage culture studies showed that the Gal-1-SLB attenuated the LPS-induced inflammation and the production of macrophage-secreted inflammatory cytokines. Finally, the implanted Gal-1-SLB reduced the infiltration of immune cells at the tissue-implant interface and increased markers for M2 polarization and blood vessel formation in vivo. This straightforward surface coating with Gal-1 can be a useful strategy for modulating the vascular and immune cells around a blood-contacting device.
Collapse
Affiliation(s)
- Md Lemon Hasan
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Ju Ro Lee
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Khandoker Asiqur Rahaman
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Dae Hyeok Yang
- Institute of Cell and Tissue Engineering, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yoon Ki Joung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
- KHU-KIST Department of Conversing Science and Technology, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Bamberg EE, Maslanka M, Vinod-Paul K, Sams S, Pollack E, Conklin M, Kabos P, Hansen KC. Obesity-driven changes in breast tissue exhibit a pro-angiogenic extracellular matrix signature. Matrix Biol Plus 2024; 24:100162. [PMID: 39380725 PMCID: PMC11460480 DOI: 10.1016/j.mbplus.2024.100162] [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: 08/07/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
Obesity has reached epidemic proportions in the United States, emerging as a risk factor for the onset of breast cancer and a harbinger of unfavorable outcomes [1], [2], [3]. Despite limited understanding of the precise mechanisms, both obesity and breast cancer are associated with extracellular matrix (ECM) rewiring [4], [5], [6]. Utilizing total breast tissue proteomics, we analyzed normal-weight (18.5 to < 25 kg/m2), overweight (25 to < 30 kg/m2), and obese (≥30 kg/m2) individuals to identify potential ECM modifying proteins for cancer development and acceleration. Obese individuals exhibited substantial ECM alterations, marked by increased basement membrane deposition, angiogenic signatures, and ECM-modifying proteins. Notably, the collagen IV crosslinking enzyme peroxidasin (PXDN) emerged as a potential mediator of the ECM changes in individuals with an elevated body mass index (BMI), strongly correlating with angiogenic and basement membrane signatures. Furthermore, glycan-binding proteins galectin-1 (LGALS1) and galectin-3 (LGALS3), which play crucial roles in matrix interactions and angiogenesis, also strongly correlate with ECM modifications. In breast cancer, elevated PXDN, LGALS1, and LGALS3 correlate with reduced relapse-free and distant-metastatic-free survival. These proteins were significantly associated with mesenchymal stromal cell markers, indicating adipocytes and fibroblasts may be the primary contributors of the obesity-related ECM changes. Our findings unveil a pro-angiogenic ECM signature in obese breast tissue, offering potential targets to inhibit breast cancer development and progression.
Collapse
Affiliation(s)
- Ellen E. Bamberg
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mark Maslanka
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kiran Vinod-Paul
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sharon Sams
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Erica Pollack
- Department of Radiology and Medical Imaging, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew Conklin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, Carbone Cancer Center (Tumor Microenvironment Program), University of Wisconsin, Madison, WI, USA
- Laboratory for Optical and Computations Instrumentation, Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Peter Kabos
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
3
|
Yang Y, Wu B, Zou W, Han F. Unveiling the molecular characteristics and antibacterial activity of tandem-repeat-type Galectin-8 in large yellow croaker (Larimichthys crocea). FISH & SHELLFISH IMMUNOLOGY 2024; 153:109849. [PMID: 39173981 DOI: 10.1016/j.fsi.2024.109849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Galectin-8 (Gal-8) is a versatile carbohydrate-binding protein with pivotal roles in immune regulation and cellular processes. This study introduces a novel galectin-8 protein, LcGal-8, from the large yellow croaker (Larimichthys crocea), showcasing typical characteristics of tandem-repeat-type galectins, including the absence of a signal peptide or transmembrane region and the presence of conserved sugar-binding motifs. Phylogenetic analysis reveals its conservation among fish species. Expression profiling indicates widespread distribution in immune tissues, particularly the spleen, implicating involvement in immune processes. The subcellular localization analysis reveals that LcGal-8 is present in both the cytoplasm and nucleus. Upon bacterial challenge, LcGal-8 is up-regulated in immune tissues, suggesting a role in host defense. Functional assays demonstrate that LcGal-8 can agglutinate gram-negative bacteria. The recombinant LcGal-8 protein agglutinates red blood cells from the large yellow croaker independently of Ca2⁺, however, this activity is inhibited by lipopolysaccharide (LPS) at 2.5 μg/mL. Fluorescence detection kits and scanning electron microscopy (SEM) confirm the agglutination and bactericidal effects of LcGal-8 against various gram-negative bacteria, including Vibrio harveyi, Aeromondaceae hydrophila, Aeromondaceae veronii, Pseudomonas plecoglossicida, Edwardsiella tarda. These findings contribute valuable insights into the genetic basis of disease resistance in the large yellow croaker and could support molecular breeding strategies to enhance disease resistance.
Collapse
Affiliation(s)
- Yao Yang
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Fisheries College, Jimei University, Xiamen, 361000, PR China
| | - Baolan Wu
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Fisheries College, Jimei University, Xiamen, 361000, PR China
| | - Wenzheng Zou
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Fisheries College, Jimei University, Xiamen, 361000, PR China
| | - Fang Han
- State Key Laboratory of Mariculture Breeding, Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-Environment, Fisheries College, Jimei University, Xiamen, 361000, PR China.
| |
Collapse
|
4
|
Ghadrdoost Nakhchi B, Kosuru R, Chrzanowska M. Towards Targeting Endothelial Rap1B to Overcome Vascular Immunosuppression in Cancer. Int J Mol Sci 2024; 25:9853. [PMID: 39337337 PMCID: PMC11432579 DOI: 10.3390/ijms25189853] [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: 07/31/2024] [Revised: 08/23/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
The vascular endothelium, a specialized monolayer of endothelial cells (ECs), is crucial for maintaining vascular homeostasis by controlling the passage of substances and cells. In the tumor microenvironment, Vascular Endothelial Growth Factor A (VEGF-A) drives tumor angiogenesis, leading to endothelial anergy and vascular immunosuppression-a state where ECs resist cytotoxic CD8+ T cell infiltration, hindering immune surveillance. Immunotherapies have shown clinical promise. However, their effectiveness is significantly reduced by tumor EC anergy. Anti-angiogenic treatments aim to normalize tumor vessels and improve immune cell infiltration. Despite their potential, these therapies often cause significant systemic toxicities, necessitating new treatments. The small GTPase Rap1B emerges as a critical regulator of Vascular Endothelial Growth Factor Receptor 2 (VEGFR2) signaling in ECs. Our studies using EC-specific Rap1B knockout mice show that the absence of Rap1B impairs tumor growth, alters vessel morphology, and increases CD8+ T cell infiltration and activation. This indicates that Rap1B mediates VEGF-A's immunosuppressive effects, making it a promising target for overcoming vascular immunosuppression in cancer. Rap1B shares structural and functional similarities with RAS oncogenes. We propose that targeting Rap1B could enhance therapies' efficacy while minimizing adverse effects by reversing endothelial anergy. We briefly discuss strategies successfully developed for targeting RAS as a model for developing anti-Rap1 therapies.
Collapse
Affiliation(s)
| | - Ramoji Kosuru
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA; (B.G.N.)
| | - Magdalena Chrzanowska
- Versiti Blood Research Institute, Milwaukee, WI 53226, USA; (B.G.N.)
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
5
|
Touarin P, Serrano B, Courbois A, Bornet O, Chen Q, Scott LG, Williamson JR, Sebban-Kreuzer C, Mancini SJC, Elantak L. Pre-B cell receptor acts as a selectivity switch for galectin-1 at the pre-B cell surface. Cell Rep 2024; 43:114541. [PMID: 39058594 DOI: 10.1016/j.celrep.2024.114541] [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: 11/22/2022] [Revised: 05/14/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Galectins are glycan-binding proteins translating the sugar-encoded information of cellular glycoconjugates into physiological activities, including immunity, cell migration, and signaling. Galectins also interact with non-glycosylated partners in the extracellular milieu, among which the pre-B cell receptor (pre-BCR) during B cell development. How these interactions might interplay with the glycan-decoding function of galectins is unknown. Here, we perform NMR experiments on native membranes to monitor Gal-1 binding to physiological cell surface ligands. We show that pre-BCR interaction changes Gal-1 binding to glycosylated pre-B cell surface receptors. At the molecular and cellular levels, we identify α2,3-sialylated motifs as key targeted epitopes. This targeting occurs through a selectivity switch increasing Gal-1 contacts with α2,3-sialylated poly-N-acetyllactosamine upon pre-BCR interaction. Importantly, we observe that this switch is involved in the regulation of pre-BCR activation. Altogether, this study demonstrates that interactions to non-glycosylated proteins regulate the glycan-decoding functions of galectins at the cell surface.
Collapse
Affiliation(s)
- Pauline Touarin
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM UMR7255), Institut de Microbiologie de la Méditerranée, Institut de Microbiologie, Bioénergies et Biotechnologies, CNRS, Aix-Marseille University, Marseille, France
| | - Bastien Serrano
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM UMR7255), Institut de Microbiologie de la Méditerranée, Institut de Microbiologie, Bioénergies et Biotechnologies, CNRS, Aix-Marseille University, Marseille, France
| | - Audrey Courbois
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM UMR7255), Institut de Microbiologie de la Méditerranée, Institut de Microbiologie, Bioénergies et Biotechnologies, CNRS, Aix-Marseille University, Marseille, France
| | - Olivier Bornet
- NMR platform, Institut de Microbiologie de la Méditerranée (IMM FR3479), Institut de Microbiologie, Bioénergies et Biotechnologies, CNRS, Aix-Marseille University, Marseille, France
| | - Qian Chen
- Cassia, 3030 Bunker Hill Street, Suite 214, San Diego, CA 92109, USA
| | - Lincoln G Scott
- Cassia, 3030 Bunker Hill Street, Suite 214, San Diego, CA 92109, USA
| | - James R Williamson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Corinne Sebban-Kreuzer
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM UMR7255), Institut de Microbiologie de la Méditerranée, Institut de Microbiologie, Bioénergies et Biotechnologies, CNRS, Aix-Marseille University, Marseille, France
| | | | - Latifa Elantak
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (LISM UMR7255), Institut de Microbiologie de la Méditerranée, Institut de Microbiologie, Bioénergies et Biotechnologies, CNRS, Aix-Marseille University, Marseille, France.
| |
Collapse
|
6
|
Thijssen VLJL. Vascular galectins in tumor angiogenesis and cancer immunity. Semin Immunopathol 2024; 46:3. [PMID: 38990363 PMCID: PMC11239785 DOI: 10.1007/s00281-024-01014-9] [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: 01/02/2024] [Accepted: 03/13/2024] [Indexed: 07/12/2024]
Abstract
Sustained tumor angiogenesis, i.e., the induction and maintenance of blood vessel growth by tumor cells, is one of the hallmarks of cancer. The vascularization of malignant tissues not only facilitates tumor growth and metastasis, but also contributes to immune evasion. Important players in all these processes are the endothelial cells which line the luminal side of blood vessel. In the tumor vasculature, these cells are actively involved in angiogenesis as well in the hampered recruitment of immune cells. This is the result of the abnormal tumor microenvironment which triggers both angiostimulatory and immune inhibitory gene expression profiles in endothelial cells. In recent years, it has become evident that galectins constitute a protein family that is expressed in the tumor endothelium. Moreover, several members of this glycan-binding protein family have been found to facilitate tumor angiogenesis and stimulate immune suppression. All this has identified galectins as potential therapeutic targets to simultaneously hamper tumor angiogenesis and alleviate immune suppression. The current review provides a brief introduction in the human galectin protein family. The current knowledge regarding the expression and regulation of galectins in endothelial cells is summarized. Furthermore, an overview of the role that endothelial galectins play in tumor angiogenesis and tumor immunomodulation is provided. Finally, some outstanding questions are discussed that should be addressed by future research efforts. This will help to fully understand the contribution of endothelial galectins to tumor progression and to exploit endothelial galectins for cancer therapy.
Collapse
Affiliation(s)
- Victor L J L Thijssen
- Radiation Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, Netherlands.
- Center for Experimental and Molecular Medicine, Laboratory for Experimental Oncology and Radiobiology, Meibergdreef 9, 1105 AZ, Amsterdam, the Netherlands.
- Cancer Center Amsterdam, Cancer Biology & Immunology, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Steffen CL, Manoharan GB, Pavic K, Yeste-Vázquez A, Knuuttila M, Arora N, Zhou Y, Härmä H, Gaigneaux A, Grossmann TN, Abankwa DK. Identification of an H-Ras nanocluster disrupting peptide. Commun Biol 2024; 7:837. [PMID: 38982284 PMCID: PMC11233548 DOI: 10.1038/s42003-024-06523-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 06/28/2024] [Indexed: 07/11/2024] Open
Abstract
Hyperactive Ras signalling is found in most cancers. Ras proteins are only active in membrane nanoclusters, which are therefore potential drug targets. We previously showed that the nanocluster scaffold galectin-1 (Gal1) enhances H-Ras nanoclustering via direct interaction with the Ras binding domain (RBD) of Raf. Here, we establish that the B-Raf preference of Gal1 emerges from the divergence of the Raf RBDs at their proposed Gal1-binding interface. We then identify the L5UR peptide, which disrupts this interaction by binding with low micromolar affinity to the B- and C-Raf-RBDs. Its 23-mer core fragment is sufficient to interfere with H-Ras nanoclustering, modulate Ras-signalling and moderately reduce cell viability. These latter two phenotypic effects may also emerge from the ability of L5UR to broadly engage with several RBD- and RA-domain containing Ras interactors. The L5UR-peptide core fragment is a starting point for the development of more specific reagents against Ras-nanoclustering and -interactors.
Collapse
Affiliation(s)
- Candy Laura Steffen
- Cancer Cell Biology and Drug Discovery group, Department of Life Sciences and Medicine, University of Luxembourg, 4362, Esch-sur-Alzette, Luxembourg
| | - Ganesh Babu Manoharan
- Cancer Cell Biology and Drug Discovery group, Department of Life Sciences and Medicine, University of Luxembourg, 4362, Esch-sur-Alzette, Luxembourg
| | - Karolina Pavic
- Cancer Cell Biology and Drug Discovery group, Department of Life Sciences and Medicine, University of Luxembourg, 4362, Esch-sur-Alzette, Luxembourg
| | - Alejandro Yeste-Vázquez
- Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), VU University Amsterdam, Amsterdam, The Netherlands
| | - Matias Knuuttila
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland
| | - Neha Arora
- Department of Integrative Biology and Pharmacology, McGovern Medical School, UT Health, Houston, TX, 77030, USA
| | - Yong Zhou
- Department of Integrative Biology and Pharmacology, McGovern Medical School, UT Health, Houston, TX, 77030, USA
| | - Harri Härmä
- Chemistry of Drug Development, Department of Chemistry, University of Turku, 20500, Turku, Finland
| | - Anthoula Gaigneaux
- Bioinformatics Core, Department of Life Sciences and Medicine, University of Luxembourg, 4367, Esch-sur-Alzette, Luxembourg
| | - Tom N Grossmann
- Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute of Molecular and Life Sciences (AIMMS), VU University Amsterdam, Amsterdam, The Netherlands
| | - Daniel Kwaku Abankwa
- Cancer Cell Biology and Drug Discovery group, Department of Life Sciences and Medicine, University of Luxembourg, 4362, Esch-sur-Alzette, Luxembourg.
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520, Turku, Finland.
| |
Collapse
|
8
|
Arvidsson D, Rodrigues Silva VR, Ekblom Ö, Ekblom-Bak E, Fryk E, Jansson PA, Börjesson M. Cardiorespiratory fitness and the association with galectin-1 in middle-aged individuals. PLoS One 2024; 19:e0301412. [PMID: 38578722 PMCID: PMC10997126 DOI: 10.1371/journal.pone.0301412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 03/16/2024] [Indexed: 04/07/2024] Open
Abstract
Galectin-1 plays a functional role in human metabolism and the levels are altered in obesity and type 2 diabetes (T2D). This study investigates the association of cardiorespiratory fitness (CRF) with galectin-1 and the interconnection with body fatness. Cross-sectional data from the Swedish CArdioPulmonary bioImage Study (SCAPIS) pilot was analyzed, including a sample of 774 middle-aged individuals. A submaximal cycle ergometer test was used to estimate CRF as an indirect measure of the physical activity (PA) level. Serum-galectin-1 concentration was determined from venous blood collected after an overnight fast. Body mass index (BMI) was used as an indirect measure of body fatness. CRF was significantly associated with galectin-1, when controlled for age and sex (regression coefficient (regr coeff) = -0.29, p<0.001). The strength of the association was attenuated when BMI was added to the regression model (regr coeff = -0.09, p = 0.07), while the association between BMI and galectin-1 remained strong (regr coeff = 0.40, p<0.001). CRF was associated with BMI (regr coeff = -0.50, p<0.001). The indirect association between CRF and galectin-1 through BMI (-0.50 x 0.40) contributed to 69% of total association (mediation analysis). In group comparisons, individuals with low CRF-high BMI had the highest mean galectin-1 level (25 ng/ml), while individuals with high CRF-low BMI had the lowest level (21 ng/ml). Intermediate levels of galectin-1 were found in the low CRF-low BMI and high CRF-high BMI groups (both 22 ng/ml). The galectin-1 level in the low CRF-high BMI group was significantly different from the other three groups (P<0.001). In conclusion, galectin-1 is associated with CRF as an indirect measure of the PA level through interconnection with body fatness. The size of the association is of clinical relevance.
Collapse
Affiliation(s)
- Daniel Arvidsson
- Center for Health and Performance, Department of Food and Nutrition, and Sport Science, Faculty of Education, University of Gothenburg, Gothenburg, Sweden
| | - Vagner Ramon Rodrigues Silva
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Örjan Ekblom
- Department of Physical Activity and Health, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Elin Ekblom-Bak
- Department of Physical Activity and Health, Swedish School of Sport and Health Sciences, Stockholm, Sweden
| | - Emanuel Fryk
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per-Anders Jansson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mats Börjesson
- Center for Lifestyle Intervention, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
| |
Collapse
|
9
|
Khan M, Cherni K, Dekhili R, Spadavecchia J. Spectroscopic Assessment of Doxorubicin (DOX)-Gemcitabine (GEM) Gold Complex Nanovector as Diagnostic Tool of Galectin-1 Biomarker. Nanotechnol Sci Appl 2024; 17:95-105. [PMID: 38567312 PMCID: PMC10986416 DOI: 10.2147/nsa.s448883] [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: 11/20/2023] [Accepted: 02/09/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction The aim of this study is focused on the development of theranostic hybrid nanovectors based on gold-doxorubicin (DOX)-gemcitabine (GEM) complexes and their active targeting with Galectin-1 (Gal-1) as a promising therapeutic and prognostic marker in cancer. Methods For this purpose, a gold salt (HAuCl4) interacts with antitumor drugs (DOX; GEM) by chelation and then stabilizes with dicarboxylic acid-terminated polyethylene glycol (PEG) as a biocompatible surfactant. The proposed methodology is fast and reproducible, and leads to the formation of a hybrid nanovector named GEM@DOX IN PEG-AuNPs, in which the chemo-biological stability was improved. All synthetic chemical products were evaluated using various spectroscopic techniques (Raman and UV-Vis spectroscopy) and transmission electron microscopy (TEM). Results To conceive a therapeutic application, our hybrid nanovector (GEM@DOX IN PEG-AuNPs) was conjugated with the Galectin-1 protein (Gal-1) at different concentrations to predict and specifically recognize cancer cells. Gal-1 interacts with GEM@DOX in PEG-AuNPs, as shown by SPR and Raman measurements. We observed both dynamic variation in the plasmon position (SPR) and Raman band with Gal-1 concentration. Discussion We identified that GEM grafted electrostatically onto DOX IN PEG-AuNPs assumes a better chemical conformation, in which the amino group (NH3+) reacts with the carboxylic (COO-) group of PEG diacide, whereas the ciclopenthanol group at position C-5' reacts with NH3+ of DOX. Conclusion This study opens further way in order to built "smart nanomedical devices" that could have a dual application as therapeutic and diagnostic in the field of nanomedicine and preclinical studies associated.
Collapse
Affiliation(s)
- Memona Khan
- CNRS, UMR 7244, NBD-CSPBAT, Laboratory of Chemistry, Structures and Properties of Biomaterials and Therapeutic Agents University Paris 13, Sorbonne Paris Nord, Bobigny, France
| | - Khaoula Cherni
- CNRS, UMR 7244, NBD-CSPBAT, Laboratory of Chemistry, Structures and Properties of Biomaterials and Therapeutic Agents University Paris 13, Sorbonne Paris Nord, Bobigny, France
| | - Rawdha Dekhili
- CNRS, UMR 7244, NBD-CSPBAT, Laboratory of Chemistry, Structures and Properties of Biomaterials and Therapeutic Agents University Paris 13, Sorbonne Paris Nord, Bobigny, France
| | - Jolanda Spadavecchia
- CNRS, UMR 7244, NBD-CSPBAT, Laboratory of Chemistry, Structures and Properties of Biomaterials and Therapeutic Agents University Paris 13, Sorbonne Paris Nord, Bobigny, France
| |
Collapse
|
10
|
Yaylim İ, Aru M, Farooqi AA, Hakan MT, Buttari B, Arese M, Saso L. Regulation of Nrf2/Keap1 signaling pathway in cancer drug resistance by galectin-1: cellular and molecular implications. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:8. [PMID: 38434765 PMCID: PMC10905161 DOI: 10.20517/cdr.2023.79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Oxidative stress is characterized by the deregulation of the redox state in the cells, which plays a role in the initiation of various types of cancers. The activity of galectin-1 (Gal-1) depends on the cell redox state and the redox state of the microenvironment. Gal-1 expression has been related to many different tumor types, as it plays important roles in several processes involved in cancer progression, such as apoptosis, cell migration, adhesion, and immune response. The erythroid-2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1) signaling pathway is a crucial mechanism involved in both cell survival and cell defense against oxidative stress. In this review, we delve into the cellular and molecular roles played by Gal-1 in the context of oxidative stress onset in cancer cells, particularly focusing on its involvement in activating the Nrf2/Keap1 signaling pathway. The emerging evidence concerning the anti-apoptotic effect of Gal-1, together with its ability to sustain the activation of the Nrf2 pathway in counteracting oxidative stress, supports the role of Gal-1 in the promotion of tumor cells proliferation, immuno-suppression, and anti-tumor drug resistance, thus highlighting that the inhibition of Gal-1 emerges as a potential strategy for the restraint and regression of tumor progression. Overall, a deeper understanding of the multi-functionality and disease-specific expression profiling of Gal-1 will be crucial for the design and development of novel Gal-1 inhibitors as anticancer agents. Excitingly, although it is still understudied, the ever-growing knowledge of the sophisticated interplay between Gal-1 and Nrf2/Keap1 will enable researchers to gain valuable insights into the underlying causes of carcinogenesis and metastasis.
Collapse
Affiliation(s)
- İlhan Yaylim
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul 34280, Turkiye
| | - Melek Aru
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul 34280, Turkiye
- Department of Medical Education, Istinye University Faculty of Medicine, Istanbul 34396, Turkiye
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan
| | - Mehmet Tolgahan Hakan
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul 34280, Turkiye
| | - Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, Rome 00161, Italy
| | - Marzia Arese
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University, Rome 00185, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, Rome 00185, Italy
| |
Collapse
|
11
|
Leone P, Malerba E, Susca N, Favoino E, Perosa F, Brunori G, Prete M, Racanelli V. Endothelial cells in tumor microenvironment: insights and perspectives. Front Immunol 2024; 15:1367875. [PMID: 38426109 PMCID: PMC10902062 DOI: 10.3389/fimmu.2024.1367875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
The tumor microenvironment is a highly complex and dynamic mixture of cell types, including tumor, immune and endothelial cells (ECs), soluble factors (cytokines, chemokines, and growth factors), blood vessels and extracellular matrix. Within this complex network, ECs are not only relevant for controlling blood fluidity and permeability, and orchestrating tumor angiogenesis but also for regulating the antitumor immune response. Lining the luminal side of vessels, ECs check the passage of molecules into the tumor compartment, regulate cellular transmigration, and interact with both circulating pathogens and innate and adaptive immune cells. Thus, they represent a first-line defense system that participates in immune responses. Tumor-associated ECs are involved in T cell priming, activation, and proliferation by acting as semi-professional antigen presenting cells. Thus, targeting ECs may assist in improving antitumor immune cell functions. Moreover, tumor-associated ECs contribute to the development at the tumor site of tertiary lymphoid structures, which have recently been associated with enhanced response to immune checkpoint inhibitors (ICI). When compared to normal ECs, tumor-associated ECs are abnormal in terms of phenotype, genetic expression profile, and functions. They are characterized by high proliferative potential and the ability to activate immunosuppressive mechanisms that support tumor progression and metastatic dissemination. A complete phenotypic and functional characterization of tumor-associated ECs could be helpful to clarify their complex role within the tumor microenvironment and to identify EC specific drug targets to improve cancer therapy. The emerging therapeutic strategies based on the combination of anti-angiogenic treatments with immunotherapy strategies, including ICI, CAR T cells and bispecific antibodies aim to impact both ECs and immune cells to block angiogenesis and at the same time to increase recruitment and activation of effector cells within the tumor.
Collapse
Affiliation(s)
- Patrizia Leone
- Internal Medicine Unit, Department of Interdisciplinary Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Eleonora Malerba
- Department of Precision and Regenerative Medicine and Ionian Area-(DiMePRe-J), Aldo Moro University of Bari, Bari, Italy
| | - Nicola Susca
- Internal Medicine Unit, Department of Interdisciplinary Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Elvira Favoino
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Federico Perosa
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Interdisciplinary Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Giuliano Brunori
- Centre for Medical Sciences, University of Trento and Nephrology and Dialysis Division, Santa Chiara Hospital, Provincial Health Care Agency (APSS), Trento, Italy
| | - Marcella Prete
- Internal Medicine Unit, Department of Interdisciplinary Medicine, Aldo Moro University of Bari, Bari, Italy
| | - Vito Racanelli
- Centre for Medical Sciences, University of Trento and Internal Medicine Division, Santa Chiara Hospital, Provincial Health Care Agency (APSS), Trento, Italy
| |
Collapse
|
12
|
Chung H, Gyu-mi P, Na YR, Lee YS, Choi H, Seok SH. Comprehensive characterization of early-programmed tumor microenvironment by tumor-associated macrophages reveals galectin-1 as an immune modulatory target in breast cancer. Theranostics 2024; 14:843-860. [PMID: 38169569 PMCID: PMC10758049 DOI: 10.7150/thno.88917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Background: In recent years, there has been considerable interest in the therapeutic targeting of tumor-associated macrophages (TAMs) to modulate the tumor microenvironment (TME), resulting in antitumoral phenotypes. However, key mediators suitable for TAM-mediated remodeling of the TME remain poorly understood. Methods: In this study, we used single-cell RNA sequencing analyses to analyze the landscape of the TME modulated by TAMs in terms of a protumor microenvironment during early tumor development. Results: Our data revealed that the depletion of TAMs leads to a decreased epithelial-to-mesenchymal transition (EMT) signature in cancer cells and a distinct transcriptional state characterized by CD8+ T cell activation. Moreover, notable alterations in gene expression were observed upon the depletion of TAMs, identifying Galectin-1 (Gal-1) as a crucial molecular factor responsible for the observed effect. Gal-1 inhibition reversed immune suppression via the reinvigoration of CD8+ T cells, impairing tumor growth and potentiating immune checkpoint inhibitors in breast tumor models. Conclusion: These results provide comprehensive insights into TAM-mediated early tumor microenvironments and reveal immune evasion mechanisms that can be targeted by Gal-1 to induce antitumor immune responses.
Collapse
Affiliation(s)
- Hyewon Chung
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 110-799, South Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center (SNUMRC), Seoul, Republic of Korea
| | - Park Gyu-mi
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 110-799, South Korea
- Department of Biomedical Sciences and Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yi Rang Na
- Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, South Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung Hyeok Seok
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 110-799, South Korea
- Department of Biomedical Sciences and Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, South Korea
| |
Collapse
|
13
|
Toudic C, Maurer M, St-Pierre G, Xiao Y, Bannert N, Lafond J, Rassart É, Sato S, Barbeau B. Galectin-1 Modulates the Fusogenic Activity of Placental Endogenous Retroviral Envelopes. Viruses 2023; 15:2441. [PMID: 38140682 PMCID: PMC10747188 DOI: 10.3390/v15122441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Syncytin-1 and -2 are glycoproteins encoded by human endogenous retrovirus (hERV) that, through their fusogenic properties, are needed for the formation of the placental syncytiotrophoblast. Previous studies suggested that these proteins, in addition to the EnvP(b) envelope protein, are also involved in other cell fusion events. Since galectin-1 is a β-galactoside-binding protein associated with cytotrophoblast fusion during placental development, we previously tested its effect on Syncytin-mediated cell fusion and showed that this protein differently modulates the fusogenic potential of Syncytin-1 and -2. Herein, we were interested in comparing the impact of galectin-1 on hERV envelope proteins in different cellular contexts. Using a syncytium assay, we first demonstrated that galectin-1 increased the fusion of Syncytin-2- and EnvP(b)-expressing cells. We then tested the infectivity of Syncytin-1 and -2 vs. VSV-G-pseudotyped viruses toward Cos-7 and various human cell lines. In the presence of galectin-1, infection of Syncytin-2-pseudotyped viruses augmented for all cell lines. In contrast, the impact of galectin-1 on the infectivity of Syncytin-1-pseudotyped viruses varied, being cell- and dose-dependent. In this study, we report the functional associations between three hERV envelope proteins and galectin-1, which should provide information on the fusogenic activity of these proteins in the placenta and other biological and pathological processes.
Collapse
Affiliation(s)
- Caroline Toudic
- Département des Sciences Biologiques and Centre d’excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (C.T.); (Y.X.); (J.L.); (É.R.)
| | - Maike Maurer
- Robert-Koch Institute, 13353 Berlin, Germany; (M.M.); (N.B.)
| | - Guillaume St-Pierre
- Glycobiology and Bioimaging Laboratory, Research Centre for Infectious Diseases and Axe Maladies Infectieuses et Immunitaires, Laval University, Quebec City, QC G1V 0A6, Canada; (G.S.-P.); (S.S.)
| | - Yong Xiao
- Département des Sciences Biologiques and Centre d’excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (C.T.); (Y.X.); (J.L.); (É.R.)
| | - Norbert Bannert
- Robert-Koch Institute, 13353 Berlin, Germany; (M.M.); (N.B.)
| | - Julie Lafond
- Département des Sciences Biologiques and Centre d’excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (C.T.); (Y.X.); (J.L.); (É.R.)
| | - Éric Rassart
- Département des Sciences Biologiques and Centre d’excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (C.T.); (Y.X.); (J.L.); (É.R.)
| | - Sachiko Sato
- Glycobiology and Bioimaging Laboratory, Research Centre for Infectious Diseases and Axe Maladies Infectieuses et Immunitaires, Laval University, Quebec City, QC G1V 0A6, Canada; (G.S.-P.); (S.S.)
| | - Benoit Barbeau
- Département des Sciences Biologiques and Centre d’excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (C.T.); (Y.X.); (J.L.); (É.R.)
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec, Montréal, QC H2X 1E3, Canada
| |
Collapse
|
14
|
Tyshkovskiy A, Ma S, Shindyapina AV, Tikhonov S, Lee SG, Bozaykut P, Castro JP, Seluanov A, Schork NJ, Gorbunova V, Dmitriev SE, Miller RA, Gladyshev VN. Distinct longevity mechanisms across and within species and their association with aging. Cell 2023; 186:2929-2949.e20. [PMID: 37269831 PMCID: PMC11192172 DOI: 10.1016/j.cell.2023.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/29/2022] [Accepted: 05/02/2023] [Indexed: 06/05/2023]
Abstract
Lifespan varies within and across species, but the general principles of its control remain unclear. Here, we conducted multi-tissue RNA-seq analyses across 41 mammalian species, identifying longevity signatures and examining their relationship with transcriptomic biomarkers of aging and established lifespan-extending interventions. An integrative analysis uncovered shared longevity mechanisms within and across species, including downregulated Igf1 and upregulated mitochondrial translation genes, and unique features, such as distinct regulation of the innate immune response and cellular respiration. Signatures of long-lived species were positively correlated with age-related changes and enriched for evolutionarily ancient essential genes, involved in proteolysis and PI3K-Akt signaling. Conversely, lifespan-extending interventions counteracted aging patterns and affected younger, mutable genes enriched for energy metabolism. The identified biomarkers revealed longevity interventions, including KU0063794, which extended mouse lifespan and healthspan. Overall, this study uncovers universal and distinct strategies of lifespan regulation within and across species and provides tools for discovering longevity interventions.
Collapse
Affiliation(s)
- Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119234, Russia
| | - Siming Ma
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anastasia V Shindyapina
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stanislav Tikhonov
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119234, Russia
| | - Sang-Goo Lee
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Perinur Bozaykut
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - José P Castro
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; Aging and Aneuploidy Laboratory, IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Andrei Seluanov
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Nicholas J Schork
- Quantitative Medicine and Systems Biology Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Sergey E Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow 119234, Russia
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
15
|
Li X, Shi W, Wei G, Lv J, Wang D, Xing B, Zhou J, Zhao J, Sun H. Galectin-1 promotes angiogenesis and chondrogenesis during antler regeneration. Cell Mol Biol Lett 2023; 28:40. [PMID: 37189051 PMCID: PMC10184426 DOI: 10.1186/s11658-023-00456-7] [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: 03/14/2023] [Accepted: 04/28/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Deer antlers are the only known mammalian structure that undergoes full regeneration. In addition, it is peculiar because when growing, it contains vascularized cartilage. The differentiation of antler stem cells (ASCs) into chondrocytes while inducing endochondral extension of blood vessels is necessary to form antler vascularized cartilage. Therefore, antlers provide an unparalleled opportunity to investigate chondrogenesis, angiogenesis, and regenerative medicine. A study found that Galectin-1 (GAL-1), which can be used as a marker in some tumors, is highly expressed in ASCs. This intrigued us to investigate what role GAL-1 could play in antler regeneration. METHODS We measured the expression level of GAL-1 in antler tissues and cells by immunohistochemistry, WB and QPCR. We constructed antlerogenic periosteal cells (APCs, one cell type of ASCs) with the GAL-1 gene knocked out (APCGAL-1-/-) using CRISPR-CAS9 gene editing system. The effect of GAL-1 on angiogenesis was determined by stimulating human umbilical vein endothelial cells (HUVECs) using APCGAL-1-/- conditioned medium or adding exogenous deer GAL-1 protein. The effect of APCGAL-1-/- on chondrogenic differentiation was evaluated compared with the APCs under micro-mass culture. The gene expression pattern of APCGAL-1-/- was analyzed by transcriptome sequencing. RESULTS Immunohistochemistry revealed that GAL-1 was widely expressed in the antlerogenic periosteum (AP), pedicle periosteum (PP) and antler growth center. Western blot and qRT-PCR analysis using deer cell lines further supports this result. The proliferation, migration, and tube formation assays of human umbilical vein endothelial cells (HUVECs) showed that the proangiogenic activity of APCGAL-1-/- medium was significantly decreased (P < 0.05) compared with the APCs medium. The proangiogenic activity of deer GAL-1 protein was further confirmed by adding exogenous deer GAL-1 protein (P < 0.05). The chondrogenic differentiation ability of APCGAL-1-/- was impeded under micro-mass culture. The terms of GO and KEGG enrichment of the differentially expressed genes (DEGs) of APCGAL-1-/- showed that down-regulated expression of pathways associated with deer antler angiogenesis, osteogenesis and stem cell pluripotency, such as the PI3K-AKT signaling pathway, signaling pathways regulating pluripotency of stem cells and TGF-β signaling pathway. CONCLUSIONS Deer GAL-1, has strong angiogenic activity, is widely and highly expressed in deer antler. The APCs can induce angiogenesis by secreting GAL-1. The knockout of GAL-1 gene of APCs damaged its ability to induce angiogenesis and differentiate into chondrocytes. This ability is crucial to the formation of deer antler vascularized cartilage. Moreover, Deer antlers offer a unique model to explore explore how angiogenesis at high levels of GAL-1 expression can be elegantly regulated without becoming cancerous.
Collapse
Affiliation(s)
- Xunsheng Li
- Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Jilin, China
| | - Wanwan Shi
- Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Jilin, China
| | - Guanning Wei
- Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Jilin, China
| | - Jinpeng Lv
- Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Jilin, China
| | - Datao Wang
- Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Jilin, China
| | - Baorui Xing
- Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Jilin, China
| | - Jue Zhou
- Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Jilin, China
| | - Jianwei Zhao
- Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Jilin, China
| | - Hongmei Sun
- Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Jilin, China.
| |
Collapse
|
16
|
Matteucci C, Nepravishta R, Argaw-Denboba A, Mandaliti W, Giovinazzo A, Petrone V, Balestrieri E, Sinibaldi-Vallebona P, Pica F, Paci M, Garaci E. Thymosin α1 interacts with Galectin-1 modulating the β-galactosides affinity and inducing alteration in the biological activity. Int Immunopharmacol 2023; 118:110113. [PMID: 37028279 DOI: 10.1016/j.intimp.2023.110113] [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: 01/31/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 04/09/2023]
Abstract
The study of mechanism of action of Thymosin alpha 1 (Tα1) and the basis of the pleiotropic effect in health and disease, is one of the main focus of our ongoing research. Tα1 is a thymic peptide that demonstrates a peculiar ability to restore homeostasis in different physiological and pathological conditions (i.e., infections, cancer, immunodeficiency, vaccination, and aging) acting as multitasking protein depending on the host state of inflammation or immune dysfunction. However, few are the information about mechanisms of action mediated by specific Tα1-target protein interaction that could explain its pleiotropic effect. We investigated the interaction of Tα1 with Galectin-1 (Gal-1), a protein belonging to an oligosaccharide binding protein family involved in a variety of biological and pathological processes, including immunoregulation, infections, cancer progression and aggressiveness. Using molecular and cellular methodological approaches, we demonstrated the interaction between these two proteins. Tα1 specifically inhibited the hemagglutination activity of Gal-1, the Gal-1 dependent in vitro formation of endothelial cell tubular structures, and the migration of cancer cells in wound healing assay. Physico-chemical methods revealed the details of the molecular interaction of Tα1 with Gal-1. Hence, the study allowed the identification of the not known until now specific interaction between Tα1 and Gal-1, and unraveled a novel mechanism of action of Tα1 that could support understanding of its pleiotropic activity.
Collapse
Affiliation(s)
- Claudia Matteucci
- Department of Experimental Medicine, University of Tor Vergata, Rome 00133, Italy.
| | - Ridvan Nepravishta
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Ayele Argaw-Denboba
- Department of Experimental Medicine, University of Tor Vergata, Rome 00133, Italy; European Molecular Biology Laboratory, EMBL, Monterotondo, Rome 00015, Italy
| | - Walter Mandaliti
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Alessandro Giovinazzo
- Department of Experimental Medicine, University of Tor Vergata, Rome 00133, Italy; Institute of Biochemistry and Cell Biology, IBBC-CNR, Monterotondo, Rome 00015, Italy
| | - Vita Petrone
- Department of Experimental Medicine, University of Tor Vergata, Rome 00133, Italy
| | - Emanuela Balestrieri
- Department of Experimental Medicine, University of Tor Vergata, Rome 00133, Italy
| | - Paola Sinibaldi-Vallebona
- Department of Experimental Medicine, University of Tor Vergata, Rome 00133, Italy; Institute of Translational Pharmacology, National Research Council, Rome 00133, Italy
| | - Francesca Pica
- Department of Experimental Medicine, University of Tor Vergata, Rome 00133, Italy
| | - Maurizio Paci
- Department of Chemical Sciences and Technologies, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Enrico Garaci
- IRCCS San Raffaele and IRCCS San Raffaele, Rome 00163, Italy; Medical and Experimental BioImaging Center, MEBIC Consortium, Rome 00166, Italy
| |
Collapse
|
17
|
Yu X, Qian J, Ding L, Yin S, Zhou L, Zheng S. Galectin-1: A Traditionally Immunosuppressive Protein Displays Context-Dependent Capacities. Int J Mol Sci 2023; 24:ijms24076501. [PMID: 37047471 PMCID: PMC10095249 DOI: 10.3390/ijms24076501] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
Galectin–Carbohydrate interactions are indispensable to pathogen recognition and immune response. Galectin-1, a ubiquitously expressed 14-kDa protein with an evolutionarily conserved β-galactoside binding site, translates glycoconjugate recognition into function. That galectin-1 is demonstrated to induce T cell apoptosis has led to substantial attention to the immunosuppressive properties of this protein, such as inducing naive immune cells to suppressive phenotypes, promoting recruitment of immunosuppressing cells as well as impairing functions of cytotoxic leukocytes. However, only in recent years have studies shown that galectin-1 appears to perform a pro-inflammatory role in certain diseases. In this review, we describe the anti-inflammatory function of galectin-1 and its possible mechanisms and summarize the existing therapies and preclinical efficacy relating to these agents. In the meantime, we also discuss the potential causal factors by which galectin-1 promotes the progression of inflammation.
Collapse
|
18
|
Ghalehbandi S, Yuzugulen J, Pranjol MZI, Pourgholami MH. The role of VEGF in cancer-induced angiogenesis and research progress of drugs targeting VEGF. Eur J Pharmacol 2023; 949:175586. [PMID: 36906141 DOI: 10.1016/j.ejphar.2023.175586] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/16/2023] [Accepted: 02/08/2023] [Indexed: 03/11/2023]
Abstract
Angiogenesis is a double-edged sword; it is a mechanism that defines the boundary between health and disease. In spite of its central role in physiological homeostasis, it provides the oxygen and nutrition needed by tumor cells to proceed from dormancy if pro-angiogenic factors tip the balance in favor of tumor angiogenesis. Among pro-angiogenic factors, vascular endothelial growth factor (VEGF) is a prominent target in therapeutic methods due to its strategic involvement in the formation of anomalous tumor vasculature. In addition, VEGF exhibits immune-regulatory properties which suppress immune cell antitumor activity. VEGF signaling through its receptors is an integral part of tumoral angiogenic approaches. A wide variety of medicines have been designed to target the ligands and receptors of this pro-angiogenic superfamily. Herein, we summarize the direct and indirect molecular mechanisms of VEGF to demonstrate its versatile role in the context of cancer angiogenesis and current transformative VEGF-targeted strategies interfering with tumor growth.
Collapse
Affiliation(s)
| | - Jale Yuzugulen
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, North Cyprus via Mersin 10, Turkey
| | | | | |
Collapse
|
19
|
Targeting galectin-driven regulatory circuits in cancer and fibrosis. Nat Rev Drug Discov 2023; 22:295-316. [PMID: 36759557 DOI: 10.1038/s41573-023-00636-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2022] [Indexed: 02/11/2023]
Abstract
Galectins are a family of endogenous glycan-binding proteins that have crucial roles in a broad range of physiological and pathological processes. As a group, these proteins use both extracellular and intracellular mechanisms as well as glycan-dependent and independent pathways to reprogramme the fate and function of numerous cell types. Given their multifunctional roles in both tissue fibrosis and cancer, galectins have been identified as potential therapeutic targets for these disorders. Here, we focus on the therapeutic relevance of galectins, particularly galectin 1 (GAL1), GAL3 and GAL9 to tumour progression and fibrotic diseases. We consider an array of galectin-targeted strategies, including small-molecule carbohydrate inhibitors, natural polysaccharides and their derivatives, peptides, peptidomimetics and biological agents (notably, neutralizing monoclonal antibodies and truncated galectins) and discuss their mechanisms of action, selectivity and therapeutic potential in preclinical models of fibrosis and cancer. We also review the results of clinical trials that aim to evaluate the efficacy of galectin inhibitors in patients with idiopathic pulmonary fibrosis, nonalcoholic steatohepatitis and cancer. The rapid pace of glycobiology research, combined with the acute need for drugs to alleviate fibrotic inflammation and overcome resistance to anticancer therapies, will accelerate the translation of anti-galectin therapeutics into clinical practice.
Collapse
|
20
|
Kapetanakis NI, Busson P. Galectins as pivotal components in oncogenesis and immune exclusion in human malignancies. Front Immunol 2023; 14:1145268. [PMID: 36817445 PMCID: PMC9935586 DOI: 10.3389/fimmu.2023.1145268] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Galectins are galactoside-binding proteins, exerting numerous functions inside and outside the cell, particularly conferring adaptation to stress factors. For most of them, aberrant expression profiles have been reported in the context of cancer. Albeit not being oncogenic drivers, galectins can be harnessed to exacerbate the malignant phenotype. Their impact on disease establishment and progression is not limited to making cancer cells resistant to apoptosis, but is prominent in the context of the tumor microenvironment, where it fosters angiogenesis, immune escape and exclusion. This review focuses mainly on Gal-1, Gal-3 and Gal-9 for which the involvement in cancer biology is best known. It presents the types of galectin dysregulations, attempts to explain the mechanisms behind them and analyzes the different ways in which they favor tumour growth. In an era where tumour resistance to immunotherapy appears as a major challenge, we highlight the crucial immunosuppressive roles of galectins and the potential therapeutic benefits of combinatorial approaches including galectin inhibition.
Collapse
Affiliation(s)
| | - Pierre Busson
- Host-Tumor Interactions in Head and Neck Carcinoma: Exploration and Therapeutic Modulations, Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche(UMR) 9018 - METabolic and SYstemic aspects of oncogenesis for new therapeutic approaches (METSY), Gustave Roussy and Université Paris-Saclay, Villejuif, France
| |
Collapse
|
21
|
Circulating galectin-1 delineates response to bevacizumab in melanoma patients and reprograms endothelial cell biology. Proc Natl Acad Sci U S A 2023; 120:e2214350120. [PMID: 36634146 PMCID: PMC9934167 DOI: 10.1073/pnas.2214350120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Blockade of vascular endothelial growth factor (VEGF) signaling with bevacizumab, a humanized anti-VEGF monoclonal antibody (mAb), or with receptor tyrosine kinase inhibitors, has improved progression-free survival and, in some indications, overall survival across several types of cancers by interrupting tumor angiogenesis. However, the clinical benefit conferred by these therapies is variable, and tumors from treated patients eventually reinitiate growth. Previously we demonstrated, in mouse tumor models, that galectin-1 (Gal1), an endogenous glycan-binding protein, preserves angiogenesis in anti-VEGF-resistant tumors by co-opting the VEGF receptor (VEGFR)2 signaling pathway in the absence of VEGF. However, the relevance of these findings in clinical settings is uncertain. Here, we explored, in a cohort of melanoma patients from AVAST-M, a multicenter, open-label, randomized controlled phase 3 trial of adjuvant bevacizumab versus standard surveillance, the role of circulating plasma Gal1 as part of a compensatory mechanism that orchestrates endothelial cell programs in bevacizumab-treated melanoma patients. We found that increasing Gal1 levels over time in patients in the bevacizumab arm, but not in the observation arm, significantly increased their risks of recurrence and death. Remarkably, plasma Gal1 was functionally active as it was able to reprogram endothelial cell biology, promoting migration, tubulogenesis, and VEGFR2 phosphorylation. These effects were prevented by blockade of Gal1 using a newly developed fully human anti-Gal1 neutralizing mAb. Thus, using samples from a large-scale clinical trial from stage II and III melanoma patients, we validated the clinical relevance of Gal1 as a potential mechanism of resistance to bevacizumab treatment.
Collapse
|
22
|
Lu ZG, Shen J, Yang J, Wang JW, Zhao RC, Zhang TL, Guo J, Zhang X. Nucleic acid drug vectors for diagnosis and treatment of brain diseases. Signal Transduct Target Ther 2023; 8:39. [PMID: 36650130 PMCID: PMC9844208 DOI: 10.1038/s41392-022-01298-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/08/2022] [Accepted: 12/21/2022] [Indexed: 01/18/2023] Open
Abstract
Nucleic acid drugs have the advantages of rich target selection, simple in design, good and enduring effect. They have been demonstrated to have irreplaceable superiority in brain disease treatment, while vectors are a decisive factor in therapeutic efficacy. Strict physiological barriers, such as degradation and clearance in circulation, blood-brain barrier, cellular uptake, endosome/lysosome barriers, release, obstruct the delivery of nucleic acid drugs to the brain by the vectors. Nucleic acid drugs against a single target are inefficient in treating brain diseases of complex pathogenesis. Differences between individual patients lead to severe uncertainties in brain disease treatment with nucleic acid drugs. In this Review, we briefly summarize the classification of nucleic acid drugs. Next, we discuss physiological barriers during drug delivery and universal coping strategies and introduce the application methods of these universal strategies to nucleic acid drug vectors. Subsequently, we explore nucleic acid drug-based multidrug regimens for the combination treatment of brain diseases and the construction of the corresponding vectors. In the following, we address the feasibility of patient stratification and personalized therapy through diagnostic information from medical imaging and the manner of introducing contrast agents into vectors. Finally, we take a perspective on the future feasibility and remaining challenges of vector-based integrated diagnosis and gene therapy for brain diseases.
Collapse
Affiliation(s)
- Zhi-Guo Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| | - Jie Shen
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Jun Yang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Jing-Wen Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Rui-Chen Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Tian-Lu Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Jing Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, P.R. China.
- University of Chinese Academy of Sciences, Beijing, 100049, P.R. China.
| |
Collapse
|
23
|
Laderach DJ, Compagno D. Inhibition of galectins in cancer: Biological challenges for their clinical application. Front Immunol 2023; 13:1104625. [PMID: 36703969 PMCID: PMC9872792 DOI: 10.3389/fimmu.2022.1104625] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Galectins play relevant roles in tumor development, progression and metastasis. Accordingly, galectins are certainly enticing targets for medical intervention in cancer. To date, however, clinical trials based on galectin inhibitors reported inconclusive results. This review summarizes the galectin inhibitors currently being evaluated and discusses some of the biological challenges that need to be addressed to improve these strategies for the benefit of cancer patients.
Collapse
Affiliation(s)
- Diego José Laderach
- Molecular and Functional Glyco-Oncology Laboratory, Instituto de Química Biológica de la Facutad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina,Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina,*Correspondence: Diego José Laderach,
| | - Daniel Compagno
- Molecular and Functional Glyco-Oncology Laboratory, Instituto de Química Biológica de la Facutad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
24
|
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: 2] [Impact Index Per Article: 2.0] [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
|
25
|
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: 1] [Impact Index Per Article: 1.0] [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
|
26
|
Capasso D, Pirone L, Di Gaetano S, Russo R, Saviano M, Frisulli V, Antonacci A, Pedone E, Scognamiglio V. Galectins detection for the diagnosis of chronic diseases: An emerging biosensor approach. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2023.116952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
27
|
Li X, Li M. The application of zebrafish patient-derived xenograft tumor models in the development of antitumor agents. Med Res Rev 2023; 43:212-236. [PMID: 36029178 DOI: 10.1002/med.21924] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/09/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023]
Abstract
The cost of antitumor drug development is enormous, yet the clinical outcomes are less than satisfactory. Therefore, it is of great importance to develop effective drug screening methods that enable accurate, rapid, and high-throughput discovery of lead compounds in the process of preclinical antitumor drug research. An effective solution is to use the patient-derived xenograft (PDX) tumor animal models, which are applicable for the elucidation of tumor pathogenesis and the preclinical testing of novel antitumor compounds. As a promising screening model organism, zebrafish has been widely applied in the construction of the PDX tumor model and the discovery of antineoplastic agents. Herein, we systematically survey the recent cutting-edge advances in zebrafish PDX models (zPDX) for studies of pathogenesis mechanisms and drug screening. In addition, the techniques used in the construction of zPDX are summarized. The advantages and limitations of the zPDX are also discussed in detail. Finally, the prospects of zPDX in drug discovery, translational medicine, and clinical precision medicine treatment are well presented.
Collapse
Affiliation(s)
- Xiang Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
28
|
Lau LS, Mohammed NBB, Dimitroff CJ. Decoding Strategies to Evade Immunoregulators Galectin-1, -3, and -9 and Their Ligands as Novel Therapeutics in Cancer Immunotherapy. Int J Mol Sci 2022; 23:15554. [PMID: 36555198 PMCID: PMC9778980 DOI: 10.3390/ijms232415554] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
Galectins are a family of ß-galactoside-binding proteins that play a variety of roles in normal physiology. In cancer, their expression levels are typically elevated and often associated with poor prognosis. They are known to fuel a variety of cancer progression pathways through their glycan-binding interactions with cancer, stromal, and immune cell surfaces. Of the 15 galectins in mammals, galectin (Gal)-1, -3, and -9 are particularly notable for their critical roles in tumor immune escape. While these galectins play integral roles in promoting cancer progression, they are also instrumental in regulating the survival, differentiation, and function of anti-tumor T cells that compromise anti-tumor immunity and weaken novel immunotherapies. To this end, there has been a surge in the development of new strategies to inhibit their pro-malignancy characteristics, particularly in reversing tumor immunosuppression through galectin-glycan ligand-targeting methods. This review examines some new approaches to evading Gal-1, -3, and -9-ligand interactions to interfere with their tumor-promoting and immunoregulating activities. Whether using neutralizing antibodies, synthetic peptides, glyco-metabolic modifiers, competitive inhibitors, vaccines, gene editing, exo-glycan modification, or chimeric antigen receptor (CAR)-T cells, these methods offer new hope of synergizing their inhibitory effects with current immunotherapeutic methods and yielding highly effective, durable responses.
Collapse
Affiliation(s)
- Lee Seng Lau
- Department of Translational Medicine, Translational Glycobiology Institute at FIU, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Norhan B. B. Mohammed
- Department of Translational Medicine, Translational Glycobiology Institute at FIU, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena 83523, Egypt
| | - Charles J. Dimitroff
- Department of Translational Medicine, Translational Glycobiology Institute at FIU, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
29
|
Godefa TM, Derks S, Thijssen VLJL. Galectins in Esophageal Cancer: Current Knowledge and Future Perspectives. Cancers (Basel) 2022; 14:5790. [PMID: 36497271 PMCID: PMC9736038 DOI: 10.3390/cancers14235790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
Esophageal cancer is a disease with poor overall survival. Despite advancements in therapeutic options, the treatment outcome of esophageal cancer patients remains dismal with an overall 5-year survival rate of approximately 20 percent. To improve treatment efficacy and patient survival, efforts are being made to identify the factors that underlie disease progression and that contribute to poor therapeutic responses. It has become clear that some of these factors reside in the tumor micro-environment. In particular, the tumor vasculature and the tumor immune micro-environment have been implicated in esophageal cancer progression and treatment response. Interestingly, galectins represent a family of glycan-binding proteins that has been linked to both tumor angiogenesis and tumor immunosuppression. Indeed, in several cancer types, galectins have been identified as diagnostic and/or prognostic markers. However, the role of galectins in esophageal cancer is still poorly understood. Here, we summarize the current literature with regard to the expression and potential functions of galectins in esophageal cancer. In addition, we highlight the gaps in the current knowledge and we propose directions for future research in order to reveal whether galectins contribute to esophageal cancer progression and provide opportunities to improve the treatment and survival of esophageal cancer patients.
Collapse
Affiliation(s)
- Tesfay M. Godefa
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology & Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, The Netherlands
| | - Sarah Derks
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology & Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, The Netherlands
| | - Victor L. J. L. Thijssen
- Cancer Center Amsterdam, Cancer Biology & Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Radiation Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
30
|
Rimal R, Desai P, Daware R, Hosseinnejad A, Prakash J, Lammers T, Singh S. Cancer-associated fibroblasts: Origin, function, imaging, and therapeutic targeting. Adv Drug Deliv Rev 2022; 189:114504. [PMID: 35998825 DOI: 10.1016/j.addr.2022.114504] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/10/2022] [Accepted: 08/17/2022] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment (TME) is emerging as one of the primary barriers in cancer therapy. Cancer-associated fibroblasts (CAF) are a common inhabitant of the TME in several tumor types and play a critical role in tumor progression and drug resistance via different mechanisms such as desmoplasia, angiogenesis, immune modulation, and cancer metabolism. Due to their abundance and significance in pro-tumorigenic mechanisms, CAF are gaining attention as a diagnostic target as well as to improve the efficacy of cancer therapy by their modulation. In this review, we highlight existing imaging techniques that are used for the visualization of CAF and CAF-induced fibrosis and provide an overview of compounds that are known to modulate CAF activity. Subsequently, we also discuss CAF-targeted and CAF-modulating nanocarriers. Finally, our review addresses ongoing challenges and provides a glimpse into the prospects that can spearhead the transition of CAF-targeted therapies from opportunity to reality.
Collapse
Affiliation(s)
- Rahul Rimal
- Max Planck Institute for Medical Research (MPImF), Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Prachi Desai
- DWI-Leibniz Institute for Interactive Materials, RWTH Aachen University, Forkenbeckstrasse 50, 52074 Aachen, Germany
| | - Rasika Daware
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Aisa Hosseinnejad
- DWI-Leibniz Institute for Interactive Materials, RWTH Aachen University, Forkenbeckstrasse 50, 52074 Aachen, Germany
| | - Jai Prakash
- Department of Advanced Organ Bioengineering and Therapeutics, Section: Engineered Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| | - Smriti Singh
- Max Planck Institute for Medical Research (MPImF), Jahnstrasse 29, 69120 Heidelberg, Germany.
| |
Collapse
|
31
|
Sanjurjo L, Broekhuizen EC, Koenen RR, Thijssen VLJL. Galectokines: The Promiscuous Relationship between Galectins and Cytokines. Biomolecules 2022; 12:1286. [PMID: 36139125 PMCID: PMC9496209 DOI: 10.3390/biom12091286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/09/2022] [Accepted: 09/10/2022] [Indexed: 11/16/2022] Open
Abstract
Galectins, a family of glycan-binding proteins, are well-known for their role in shaping the immune microenvironment. They can directly affect the activity and survival of different immune cell subtypes. Recent evidence suggests that galectins also indirectly affect the immune response by binding to members of another immunoregulatory protein family, i.e., cytokines. Such galectin-cytokine heterodimers, here referred to as galectokines, add a new layer of complexity to the regulation of immune homeostasis. Here, we summarize the current knowledge with regard to galectokine formation and function. We describe the known and potential mechanisms by which galectokines can help to shape the immune microenvironment. Finally, the outstanding questions and challenges for future research regarding the role of galectokines in immunomodulation are discussed.
Collapse
Affiliation(s)
- Lucía Sanjurjo
- Health Research Institute of Santiago de Compostela (IDIS), Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Barcelona Ave., 15782 Santiago de Compostela, Spain
| | - Esmee C. Broekhuizen
- Department of Radiation Oncology, Amsterdam UMC Location VUmc, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Rory R. Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Victor L. J. L. Thijssen
- Department of Radiation Oncology, Amsterdam UMC Location VUmc, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology & Immunology, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
32
|
Chen M, Shi JL, Zheng ZM, Lin Z, Li MQ, Shao J. Galectins: Important Regulators in Normal and Pathologic Pregnancies. Int J Mol Sci 2022; 23:ijms231710110. [PMID: 36077508 PMCID: PMC9456357 DOI: 10.3390/ijms231710110] [Citation(s) in RCA: 3] [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: 08/05/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Galectins (Gal) are characterized by their affinity for galactoside structures on glycoconjugates. This relationship is mediated by carbohydrate recognition domains, which are multifunctional regulators of basic cellular biological processes with high structural similarity among family members. They participate in both innate and adaptive immune responses, as well as in reproductive immunology. Recently, the discovery that galectins are highly expressed at the maternal–fetal interface has garnerd the interest of experts in human reproduction. Galectins are involved in a variety of functions such as maternal–fetal immune tolerance, angiogenesis, trophoblast invasion and placental development and are considered to be important mediators of successful embryo implantation and during pregnancy. Dysregulation of these galectins is associated with abnormal and pathological pregnancies (e.g., preeclampsia, gestational diabetes mellitus, fetal growth restriction, preterm birth). Our work reviews the regulatory mechanisms of galectins in normal and pathological pregnancies and has implications for clinicians in the prevention, diagnosis and treatment of pregnancy-related diseases.
Collapse
Affiliation(s)
- Min Chen
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China
| | - Jia-Lu Shi
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China
| | - Zi-Meng Zheng
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China
| | - Zhi Lin
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China
- NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Fudan University, Shanghai 201203, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, China
- Correspondence: (M.-Q.L.); (J.S.)
| | - Jun Shao
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, China
- Department of Obstetrics, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200011, China
- Correspondence: (M.-Q.L.); (J.S.)
| |
Collapse
|
33
|
Chen Y, Liu Y, Zhang Y, Yu J, Tang L. Galectin-1 deletion in mice causes bone loss via impaired osteogenic differentiation potential of BMSCs. FASEB J 2022; 36:e22516. [PMID: 36006656 DOI: 10.1096/fj.202200397r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/11/2022] [Accepted: 08/15/2022] [Indexed: 11/11/2022]
Abstract
Bone formation is dependent on the osteoblasts which are differentiated from bone marrow stromal cells (BMSCs). In addition to potent proliferation, self-renewal, and pluripotent differentiation, BMSCs have been extensively studied due to their low immunogenicity and immunomodulatory effects. Recently, galectin-1 (Gal-1) has been proposed as a potent mediator of immunomodulatory properties of BMSCs. Previous study demonstrated that Gal-1 showed age-related decline in mice serum and serum Gal-1 was positively associated with bone mass in mice. The current study makes attempts to elucidate the functional role of Gal-1 in skeletal system by investigating the regulation of Gal-1 expression during BMSCs osteogenic differentiation and the molecular mechanisms underlying the effects of Gal-1 on BMSCs osteogenic differentiation. In Gal-1 null (-/-) mice, bone loss was observed due to bone formation attenuation. In in vitro experiments, Gal-1 supported the osteogenic differentiation of BMSCs by binding to CD146 to activate Lrp5 expression and Wnt/β-catenin signaling pathway. Meanwhile, there was positive feedback regulation via Wnt/β-catenin signaling to maintain Gal-1 high-level expression during osteogenic differentiation of BMSCs. More importantly, Gal-1 down-regulation in BMSCs and attenuation of osteogenic differentiation potential of BMSCs were observed in aged mice compared with young mice. Gal-1 over-expression could enhance osteogenic differentiation potential of aged BMSCs. Our study will benefit not only for deeper insights into the functional role of Gal-1 but also for finding new targets to modulate BMSCs osteogenic differentiation.
Collapse
Affiliation(s)
- Yu Chen
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yanbin Liu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Yangyang Zhang
- Department of Orthopedic Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jiangming Yu
- Department of Orthopedic Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Liang Tang
- Department of Orthopedic Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
34
|
Vaccination against galectin-1 promotes cytotoxic T-cell infiltration in melanoma and reduces tumor burden. Cancer Immunol Immunother 2022; 71:2029-2040. [PMID: 35018481 PMCID: PMC9293851 DOI: 10.1007/s00262-021-03139-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 12/29/2021] [Indexed: 11/14/2022]
Abstract
Galectin-1 (Gal1) is a glycan-binding protein that promotes tumor progression by several distinct mechanisms. Through direct binding to vascular endothelial growth factor (VEGF)-receptor 2, Gal1 is able to induce VEGF-like signaling, which contributes to tumor angiogenesis. Furthermore, several studies have demonstrated an immunosuppressive function of Gal1 through effects on both effector and regulatory T cells. Elevated Gal1 expression and secretion have been shown in many tumor types, and high Gal1 serum levels have been connected to poor prognosis in cancer patients. These findings suggest that therapeutic strategies directed against Gal1 would enable simultaneous targeting of angiogenesis, immune evasion and metastasis. In the current study, we have analyzed the potential of Gal1 as a cancer vaccine target. We show that it is possible to generate high anti-Gal1 antibody levels in mice immunized with a recombinant vaccine protein consisting of bacterial sequences fused to Gal1. Growth of Gal1 expressing melanomas was significantly impaired in the immunized mice compared to the control group. This was associated with improved perfusion of the tumor vasculature, as well as increased infiltration of macrophages and cytotoxic T cells (CTLs). The level of granzyme B, mainly originating from CTLs in our model, was significantly elevated in Gal1 vaccinated mice and correlated with a decrease in tumor burden. We conclude that vaccination against Gal1 is a promising pro-immunogenic approach for cancer therapy that could potentially enhance the effect of other immunotherapeutic strategies due to its ability to promote CTL influx in tumors.
Collapse
|
35
|
Stroes CI, Schokker S, Khurshed M, van der Woude SO, Mathôt RA, Slingerland M, de Vos-Geelen J, Zucchetti M, Matteo C, van Dijk E, Ylstra B, Thijssen V, Derks S, Godefa T, Dijksterhuis W, Breimer GE, van Delden OM, Verhoeven RH, Meijer SL, Bijlsma MF, van Laarhoven HW. A phase Ib/II study of regorafenib and paclitaxel in patients with beyond first-line advanced esophagogastric carcinoma (REPEAT). Ther Adv Med Oncol 2022; 14:17588359221109196. [PMID: 35782751 PMCID: PMC9244942 DOI: 10.1177/17588359221109196] [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: 01/20/2022] [Accepted: 06/06/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose: Regorafenib monotherapy, a multikinase inhibitor of angiogenesis, tumor microenvironment, and tumorigenesis, showed promising results in gastric cancer. We aimed to assess the tolerability of regorafenib and paclitaxel in patients with advanced esophagogastric cancer (EGC) refractory to first-line treatment, and explore potential biomarkers. Methods: Patients received paclitaxel (80 mg/m2) on days 1, 8, and 15 of a 28-day cycle and regorafenib (80/120/160 mg) on days 1–21 in the dose-escalation cohort, and the maximum-tolerated dose (MTD) in the dose-expansion cohort. Exploratory, overall survival (OS) and progression-free survival (PFS) were compared to a propensity-score matched cohort receiving standard second-/third-line systemic treatment. Paclitaxel pharmacokinetics were assessed using samples from day 1 (D1) and day 15 (D15). We performed enzyme-linked immunosorbent assay measurements of galectin-1, RNA sequencing, and shallow whole-genome sequencing of metastatic tumor biopsies for biomarker analyses. Results: In the dose-escalation cohort (n = 14), the MTD of regorafenib was 120 mg. In all, 34 patients were enrolled in the dose-expansion cohort. Most common toxicities (all grades; grade ⩾ 3) were fatigue (79%; 4%) and sensory neuropathy (63%; 4%). Best responses achieved were partial response (28%) and stable disease (54%). Median OS and PFS were 7.8 and 4.2 months, respectively (median follow-up: 7.8 months). OS (p = 0.08) and PFS (p = 0.81) were not significantly improved compared to the matched cohort. Paclitaxel concentrations were significantly increased with regorafenib (D15) compared with paclitaxel only (D1; p < 0.05); no associations were observed with toxicity or efficacy. An increase in circulating galectin-1 compared to baseline was associated with shorter OS (p < 0.01). Enrichment of angiogenesis-related gene expression was observed in short survivors measured by RNA sequencing. Chromosome 19q13.12-q13.2 amplification was associated with shorter OS (p = 0.02) and PFS (p = 0.02). Conclusion: Treatment with regorafenib and paclitaxel is tolerable and shows promising efficacy in advanced EGC refractory to first-line treatment. Galectin-1 and chromosome 19q13.12-q13.2 amplification could serve as negative predictive biomarkers for treatment response. Registration: Clinicaltrials.gov, NCT02406170, https://clinicaltrials.gov/ct2/show/NCT02406170
Collapse
Affiliation(s)
- Charlotte I Stroes
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Sandor Schokker
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Mohammed Khurshed
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Stephanie O van der Woude
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Ron Aa Mathôt
- Department of Clinical Pharmacology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marije Slingerland
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Judith de Vos-Geelen
- Department of Internal Medicine, Division of Medical Oncology, GROW-School for Oncology and Development Biology, Maastricht UMC+, Maastricht, The Netherlands
| | - Massimo Zucchetti
- Department of Oncology, Laboratory of Cancer Pharmacology, Istituto di Ricerche Farmacologice Mario Negri IRCCS, Milan, Italy
| | - Cristina Matteo
- Department of Oncology, Laboratory of Cancer Pharmacology, Istituto di Ricerche Farmacologice Mario Negri IRCCS, Milan, Italy
| | - Erik van Dijk
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Bauke Ylstra
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Victor Thijssen
- Department of Radiation Oncology, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Sarah Derks
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Tesfay Godefa
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Willemieke Dijksterhuis
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Gerben E Breimer
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Otto M van Delden
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Rob Ha Verhoeven
- Department of Research, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, The Netherlands
| | - Sybren L Meijer
- Department of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Maarten F Bijlsma
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Hanneke Wm van Laarhoven
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Cancer Center Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| |
Collapse
|
36
|
Guda MR, Tsung AJ, Asuthkar S, Velpula KK. Galectin-1 activates carbonic anhydrase IX and modulates glioma metabolism. Cell Death Dis 2022; 13:574. [PMID: 35773253 PMCID: PMC9247167 DOI: 10.1038/s41419-022-05024-z] [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: 02/09/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 01/21/2023]
Abstract
Galectins are a family of β-galactose-specific binding proteins residing within the cytosol or nucleus, with a highly conserved carbohydrate recognition domain across many species. Accumulating evidence shows that Galectin 1 (Gal-1) plays an essential role in cancer, and its expression correlates with tumor aggressiveness and progression. Our preliminary data showed Gal-1 promotes glioma stem cell (GSC) growth via increased Warburg effect. mRNA expression and clinical data were obtained from The Cancer Genome Atlas database. The immunoblot analysis conducted using our cohort of human glioblastoma patient specimens (hGBM), confirmed Gal-1 upregulation in GBM. GC/MS analysis to evaluate the effects of Gal-1 depletion showed elevated levels of α-ketoglutaric acid, and citric acid with a concomitant reduction in lactic acid levels. Using Biolog microplate-1 mitochondrial functional assay, we confirmed that the depletion of Gal-1 increases the expression levels of the enzymes from the TCA cycle, suggesting a reversal of the Warburg phenotype. Manipulation of Gal-1 using RNA interference showed reduced ATP, lactate levels, cell viability, colony-forming abilities, and increased expression levels of genes implicated in the induction of apoptosis. Gal-1 exerts its metabolic role via regulating the expression of carbonic anhydrase IX (CA-IX), a surrogate marker for hypoxia. CA-IX functions downstream to Gal-1, and co-immunoprecipitation experiments along with proximity ligation assays confirm that Gal-1 physically associates with CA-IX to regulate its expression. Further, silencing of Gal-1 in mice models showed reduced tumor burden and increased survival compared to the mice implanted with GSC controls. Further investigation of Gal-1 in GSC progression and metabolic reprogramming is warranted.
Collapse
Affiliation(s)
- Maheedhara R. Guda
- grid.430852.80000 0001 0741 4132Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL USA
| | - Andrew J. Tsung
- grid.430852.80000 0001 0741 4132Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL USA ,grid.430852.80000 0001 0741 4132Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL USA ,Illinois Neurological Institute, Peoria, IL USA
| | - Swapna Asuthkar
- grid.430852.80000 0001 0741 4132Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL USA
| | - Kiran K. Velpula
- grid.430852.80000 0001 0741 4132Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL USA ,grid.430852.80000 0001 0741 4132Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL USA ,grid.430852.80000 0001 0741 4132Department of Pediatrics, University of Illinois College of Medicine at Peoria, Peoria, IL USA
| |
Collapse
|
37
|
Systematic Review: Targeted Molecular Imaging of Angiogenesis and Its Mediators in Rheumatoid Arthritis. Int J Mol Sci 2022; 23:ijms23137071. [PMID: 35806074 PMCID: PMC9267012 DOI: 10.3390/ijms23137071] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 12/14/2022] Open
Abstract
Extensive angiogenesis is a characteristic feature in the synovial tissue of rheumatoid arthritis (RA) from a very early stage of the disease onward and constitutes a crucial event for the development of the proliferative synovium. This process is markedly intensified in patients with prolonged disease duration, high disease activity, disease severity, and significant inflammatory cell infiltration. Angiogenesis is therefore an interesting target for the development of new therapeutic approaches as well as disease monitoring strategies in RA. To this end, nuclear imaging modalities represent valuable non-invasive tools that can selectively target molecular markers of angiogenesis and accurately and quantitatively track molecular changes in multiple joints simultaneously. This systematic review summarizes the imaging markers used for single photon emission computed tomography (SPECT) and/or positron emission tomography (PET) approaches, targeting pathways and mediators involved in synovial neo-angiogenesis in RA.
Collapse
|
38
|
Extracellular vimentin mimics VEGF and is a target for anti-angiogenic immunotherapy. Nat Commun 2022; 13:2842. [PMID: 35606362 PMCID: PMC9126915 DOI: 10.1038/s41467-022-30063-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
Anti-angiogenic cancer therapies possess immune-stimulatory properties by counteracting pro-angiogenic molecular mechanisms. We report that tumor endothelial cells ubiquitously overexpress and secrete the intermediate filament protein vimentin through type III unconventional secretion mechanisms. Extracellular vimentin is pro-angiogenic and functionally mimics VEGF action, while concomitantly acting as inhibitor of leukocyte-endothelial interactions. Antibody targeting of extracellular vimentin shows inhibition of angiogenesis in vitro and in vivo. Effective and safe inhibition of angiogenesis and tumor growth in several preclinical and clinical studies is demonstrated using a vaccination strategy against extracellular vimentin. Targeting vimentin induces a pro-inflammatory condition in the tumor, exemplified by induction of the endothelial adhesion molecule ICAM1, suppression of PD-L1, and altered immune cell profiles. Our findings show that extracellular vimentin contributes to immune suppression and functions as a vascular immune checkpoint molecule. Targeting of extracellular vimentin presents therefore an anti-angiogenic immunotherapy strategy against cancer. The pro-tumorigenic effects of vimentin have been attributed to intracellular functions in tumour cells so far. Here, the authors show that tumour endothelial cells can secrete vimentin as a pro-angiogenic factor and that targeting of vimentin can be used as an immunotherapeutic strategy.
Collapse
|
39
|
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
|
40
|
Hermenean A, Oatis D, Herman H, Ciceu A, D’Amico G, Trotta MC. Galectin 1-A Key Player between Tissue Repair and Fibrosis. Int J Mol Sci 2022; 23:ijms23105548. [PMID: 35628357 PMCID: PMC9142121 DOI: 10.3390/ijms23105548] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 12/20/2022] Open
Abstract
Galectins are ten family members of carbohydrate-binding proteins with a high affinity for β galactose-containing oligosaccharides. Galectin-1 (Gal-1) is the first protein discovered in the family, expressed in many sites under normal and pathological conditions. In the first part of the review article, we described recent advances in the Gal-1 modulatory role on wound healing, by focusing on the different phases triggered by Gal-1, such as inflammation, proliferation, tissue repair and re-epithelialization. On the contrary, Gal-1 persistent over-expression enhances angiogenesis and extracellular matrix (ECM) production via PI3K/Akt pathway activation and leads to keloid tissue. Therefore, the targeted Gal-1 modulation should be considered a method of choice to treat wound healing and avoid keloid formation. In the second part of the review article, we discuss studies clarifying the role of Gal-1 in the pathogenesis of proliferative diabetic retinopathy, liver, renal, pancreatic and pulmonary fibrosis. This evidence suggests that Gal-1 may become a biomarker for the diagnosis and prognosis of tissue fibrosis and a promising molecular target for the development of new and original therapeutic tools to treat fibrosis in different chronic diseases.
Collapse
Affiliation(s)
- Anca Hermenean
- Faculty of Medicine, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310414 Arad, Romania; (H.H.); (A.C.); (G.D.)
- Correspondence:
| | - Daniela Oatis
- Faculty of Medicine, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
| | - Hildegard Herman
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310414 Arad, Romania; (H.H.); (A.C.); (G.D.)
| | - Alina Ciceu
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310414 Arad, Romania; (H.H.); (A.C.); (G.D.)
| | - Giovanbattista D’Amico
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310414 Arad, Romania; (H.H.); (A.C.); (G.D.)
| | - Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| |
Collapse
|
41
|
Hillenmayer A, Wertheimer CM, Geerlof A, Eibl KH, Priglinger S, Priglinger C, Ohlmann A. Galectin-1 and -3 in high amounts inhibit angiogenic properties of human retinal microvascular endothelial cells in vitro. PLoS One 2022; 17:e0265805. [PMID: 35320287 PMCID: PMC8942239 DOI: 10.1371/journal.pone.0265805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/08/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose Galectin-1 and -3 are β-galactoside binding lectins with varying effects on angiogenesis and apoptosis. Since in retinal pigment epithelial cells high amounts of human recombinant galectin (hr-GAL)1 and 3 inhibit cell adhesion, migration and proliferation, we investigated if hr-GAL1 and 3 have homologous effects on human retinal microvascular endothelial cells (HRMEC) in vitro. Methods To investigate the effect of galectin-1 and -3 on HRMEC, proliferation, apoptosis and viability were analyzed after incubation with 30, 60 and 120 μg/ml hr-GAL1 or 3 by BrdU-ELISA, histone-DNA complex ELISA, live/dead staining and the WST-1 assay, respectively. Further on, a cell adhesion as well as tube formation assay were performed on galectin-treated HRMEC. Migration was investigated by the scratch migration assay and time-lapse microscopy. In addition, immunohistochemical staining on HRMEC for β-catenin, galectin-1 and -3 were performed and β-catenin expression was investigated by western blot analysis. Results Incubation with hr-GAL1 or 3 lead to a decrease in proliferation, migration, adhesion and tube formation of HRMEC compared to the untreated controls. No toxic effects of hr-GAL1 and 3 on HRMEC were detected. Intriguingly, after treatment of HRMEC with hr-GAL1 or 3, an activation of the proangiogenic Wnt/β-catenin signaling pathway was observed. However, incubation of HRMEC with hr-GAL1 or 3 drew intracellular galectin-1 and -3 out of the cells, respectively. Conclusion Exogenously added hr-GAL1 or 3 inhibit angiogenic properties of HRMEC in vitro, an effect that might be mediated via a loss of intracellular endogenous galectins.
Collapse
Affiliation(s)
- Anna Hillenmayer
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Germany
- Department of Ophthalmology, University Hospital Ulm, Ulm, Germany
| | - Christian M. Wertheimer
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Germany
- Department of Ophthalmology, University Hospital Ulm, Ulm, Germany
| | - Arie Geerlof
- Protein Expression and Purification Facility, Institute of Structural Biology, Helmholtz Center Munich for Environmental Health, Neuherberg, Germany
| | - Kirsten H. Eibl
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Germany
| | | | - Claudia Priglinger
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Germany
| | - Andreas Ohlmann
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Germany
- * E-mail:
| |
Collapse
|
42
|
Vang S, Cochran P, Sebastian Domingo J, Krick S, Barnes JW. The Glycobiology of Pulmonary Arterial Hypertension. Metabolites 2022; 12:metabo12040316. [PMID: 35448503 PMCID: PMC9026683 DOI: 10.3390/metabo12040316] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive pulmonary vascular disease of complex etiology. Cases of PAH that do not receive therapy after diagnosis have a low survival rate. Multiple reports have shown that idiopathic PAH, or IPAH, is associated with metabolic dysregulation including altered bioavailability of nitric oxide (NO) and dysregulated glucose metabolism. Multiple processes such as increased proliferation of pulmonary vascular cells, angiogenesis, apoptotic resistance, and vasoconstriction may be regulated by the metabolic changes demonstrated in PAH. Recent reports have underscored similarities between metabolic abnormalities in cancer and IPAH. In particular, increased glucose uptake and altered glucose utilization have been documented and have been linked to the aforementioned processes. We were the first to report a link between altered glucose metabolism and changes in glycosylation. Subsequent reports have highlighted similar findings, including a potential role for altered metabolism and aberrant glycosylation in IPAH pathogenesis. This review will detail research findings that demonstrate metabolic dysregulation in PAH with an emphasis on glycobiology. Furthermore, this report will illustrate the similarities in the pathobiology of PAH and cancer and highlight the novel findings that researchers have explored in the field.
Collapse
|
43
|
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: 24] [Impact Index Per Article: 12.0] [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
|
44
|
Huijbers EJM, Khan KA, Kerbel RS, Griffioen AW. Tumors resurrect an embryonic vascular program to escape immunity. Sci Immunol 2022; 7:eabm6388. [PMID: 35030032 DOI: 10.1126/sciimmunol.abm6388] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Elisabeth J M Huijbers
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Kabir A Khan
- Biological Sciences Platform, Sunnybrook Research Institute, Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Robert S Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
45
|
Bannoud N, García PA, Gambarte-Tudela J, Sundblad V, Cagnoni AJ, Bach CA, Pérez Saez JM, Blidner AG, Maller SM, Mariño KV, Salatino M, Cerliani JP, Rabinovich GA, Croci DO. Untangling Galectin-Mediated Circuits that Control Hypoxia-Driven Angiogenesis. Methods Mol Biol 2022; 2442:635-653. [PMID: 35320550 DOI: 10.1007/978-1-0716-2055-7_34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Development of an aberrant vascular network is a hallmark of the multistep pathological process of tumor growth and metastasis. In response to hypoxia, several pro-angiogenic factors are synthesized to support vascularization programs required for cancer progression. Emerging data indicate the involvement of glycans and glycan-binding proteins as critical regulators of vascular circuits in health and disease. Galectins may be regulated by hypoxic conditions and control angiogenesis in different physiopathological settings. These β-galactoside-binding proteins may promote sprouting angiogenesis by interacting with different glycosylated receptors and triggering distinct signaling pathways. Understanding the role of galectins in tumor neovascularization will contribute to the design of novel anti-angiogenic therapies aimed at complementing current anti-cancer modalities and overcoming resistance to these treatments. Here we describe selected strategies and methods used to study the role of hypoxia-regulated galectins in the regulation of blood vessel formation.
Collapse
Affiliation(s)
- Nadia Bannoud
- Instituto de Histología y Embriología de Mendoza (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - P Alfredo García
- Instituto de Histología y Embriología de Mendoza (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Julian Gambarte-Tudela
- Instituto de Histología y Embriología de Mendoza (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Victoria Sundblad
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Alejandro J Cagnoni
- Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Camila A Bach
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Juan M Pérez Saez
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ada G Blidner
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Sebastián M Maller
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Karina V Mariño
- Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mariana Salatino
- 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
| | - 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
| | - Diego O Croci
- Instituto de Histología y Embriología de Mendoza (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina.
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina.
| |
Collapse
|
46
|
Castricum KCM, Thijssen VLJL. Method to Study the Role of Galectins in Angiogenesis In Vivo Using the Chick Chorioallantoic Membrane Assay. Methods Mol Biol 2022; 2442:621-633. [PMID: 35320549 DOI: 10.1007/978-1-0716-2055-7_33] [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
Angiogenesis is a complex multi-step process involving various activities of endothelial cells. These activities are influenced in vivo by environmental conditions like interactions with other cell types and the microenvironment. Galectins play a role in several of these interactions and are therefore required for proper execution of in vivo angiogenesis. This chapter describes a method to study galectins during physiologic and pathophysiologic angiogenesis in vivo using the chicken chorioallantoic membrane (CAM) assay.
Collapse
Affiliation(s)
- Kitty C M Castricum
- Amsterdam UMC location VUmc, Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Victor L J L Thijssen
- Amsterdam UMC location VUmc, Department of Radiation Oncology, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
47
|
Videla-Richardson GA, Morris-Hanon O, Torres NI, Esquivel MI, Vera MB, Ripari LB, Croci DO, Sevlever GE, Rabinovich GA. Galectins as Emerging Glyco-Checkpoints and Therapeutic Targets in Glioblastoma. Int J Mol Sci 2021; 23:ijms23010316. [PMID: 35008740 PMCID: PMC8745137 DOI: 10.3390/ijms23010316] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 02/08/2023] Open
Abstract
Despite recent advances in diagnosis and treatment, glioblastoma (GBM) represents the most common and aggressive brain tumor in the adult population, urging identification of new rational therapeutic targets. Galectins, a family of glycan-binding proteins, are highly expressed in the tumor microenvironment (TME) and delineate prognosis and clinical outcome in patients with GBM. These endogenous lectins play key roles in different hallmarks of cancer by modulating tumor cell proliferation, oncogenic signaling, migration, vascularization and immunity. Additionally, they have emerged as mediators of resistance to different anticancer treatments, including chemotherapy, radiotherapy, immunotherapy, and antiangiogenic therapy. Particularly in GBM, galectins control tumor cell transformation and proliferation, reprogram tumor cell migration and invasion, promote vascularization, modulate cell death pathways, and shape the tumor-immune landscape by targeting myeloid, natural killer (NK), and CD8+ T cell compartments. Here, we discuss the role of galectins, particularly galectin-1, -3, -8, and -9, as emerging glyco-checkpoints that control different mechanisms associated with GBM progression, and discuss possible therapeutic opportunities based on inhibition of galectin-driven circuits, either alone or in combination with other treatment modalities.
Collapse
Affiliation(s)
- Guillermo A. Videla-Richardson
- Laboratorio de Investigación Aplicada en Neurociencias (LIAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Belén de Escobar B1625, Argentina; (G.A.V.-R.); (O.M.-H.); (M.I.E.); (M.B.V.); (L.B.R.); (G.E.S.)
| | - Olivia Morris-Hanon
- Laboratorio de Investigación Aplicada en Neurociencias (LIAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Belén de Escobar B1625, Argentina; (G.A.V.-R.); (O.M.-H.); (M.I.E.); (M.B.V.); (L.B.R.); (G.E.S.)
| | - Nicolás I. Torres
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina;
| | - Myrian I. Esquivel
- Laboratorio de Investigación Aplicada en Neurociencias (LIAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Belén de Escobar B1625, Argentina; (G.A.V.-R.); (O.M.-H.); (M.I.E.); (M.B.V.); (L.B.R.); (G.E.S.)
| | - Mariana B. Vera
- Laboratorio de Investigación Aplicada en Neurociencias (LIAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Belén de Escobar B1625, Argentina; (G.A.V.-R.); (O.M.-H.); (M.I.E.); (M.B.V.); (L.B.R.); (G.E.S.)
| | - Luisina B. Ripari
- Laboratorio de Investigación Aplicada en Neurociencias (LIAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Belén de Escobar B1625, Argentina; (G.A.V.-R.); (O.M.-H.); (M.I.E.); (M.B.V.); (L.B.R.); (G.E.S.)
| | - Diego O. Croci
- Laboratorio de Inmunopatología, Instituto de Histología y Embriología de Mendoza (IHEM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza C5500, Argentina;
| | - Gustavo E. Sevlever
- Laboratorio de Investigación Aplicada en Neurociencias (LIAN), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Belén de Escobar B1625, Argentina; (G.A.V.-R.); (O.M.-H.); (M.I.E.); (M.B.V.); (L.B.R.); (G.E.S.)
| | - Gabriel A. Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina;
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428, Argentina
- Correspondence: ; Tel.: +54-11-4783-2869 (ext. 266)
| |
Collapse
|
48
|
Chemokines modulate glycan binding and the immunoregulatory activity of galectins. Commun Biol 2021; 4:1415. [PMID: 34931005 PMCID: PMC8688422 DOI: 10.1038/s42003-021-02922-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 11/25/2021] [Indexed: 12/29/2022] Open
Abstract
Galectins are versatile glycan-binding proteins involved in immunomodulation. Evidence suggests that galectins can control the immunoregulatory function of cytokines and chemokines through direct binding. Here, we report on an inverse mechanism in which chemokines control the immunomodulatory functions of galectins. We show the existence of several specific galectin-chemokine binding pairs, including galectin-1/CXCL4. NMR analyses show that CXCL4 binding induces changes in the galectin-1 carbohydrate binding site. Consequently, CXCL4 alters the glycan-binding affinity and specificity of galectin-1. Regarding immunomodulation, CXCL4 significantly increases the apoptotic activity of galectin-1 on activated CD8+ T cells, while no effect is observed in CD4+ T cells. The opposite is found for another galectin-chemokine pair, i.e., galectin-9/CCL5. This heterodimer significantly reduces the galectin-9 induced apoptosis of CD4+ T cells and not of CD8+ T cells. Collectively, the current study describes an immunomodulatory mechanism in which specific galectin-chemokine interactions control the glycan-binding activity and immunoregulatory function of galectins.
Collapse
|
49
|
Detoxification, Hydrogen Sulphide Metabolism and Wound Healing Are the Main Functions That Differentiate Caecum Protein Expression from Ileum of Week-Old Chicken. Animals (Basel) 2021; 11:ani11113155. [PMID: 34827887 PMCID: PMC8614574 DOI: 10.3390/ani11113155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Although the ileum and caecum represent adjacent parts of the gastrointestinal tract, both compartments differ by function as well as inner environment parameters such as oxygen availability or density of colonising microbiota. As the function of a particular tissue is generally reflected by protein expression, mass spectrometry proteomics was used to characterise expressed proteins of both segments of the gastrointestinal tract. Differentially expressed proteins were identified and grouped according to biological processes specific to both gut compartments. Abstract Sections of chicken gut differ in many aspects, e.g., the passage of digesta (continuous vs. discontinuous), the concentration of oxygen, and the density of colonising microbiota. Using an unbiased LC-MS/MS protocol, we compared protein expression in 18 ileal and 57 caecal tissue samples that originated from 7-day old ISA brown chickens. We found that proteins specific to the ileum were either structural (e.g., 3 actin isoforms, villin, or myosin 1A), or those required for nutrient digestion (e.g., sucrose isomaltase, maltase–glucoamylase, peptidase D) and absorption (e.g., fatty acid-binding protein 2 and 6 or bile acid–CoA:amino acid N-acyltransferase). On the other hand, proteins characteristic of the caecum were involved in sensing and limiting the consequences of oxidative stress (e.g., thioredoxin, peroxiredoxin 6), cell adhesion, and motility associated with wound healing (e.g., fibronectin 1, desmoyokin). These mechanisms are coupled with the activation of mechanisms suppressing the inflammatory response (galectin 1). Rather prominent were also expressions of proteins linked to hydrogen sulphide metabolism in caecum represented by cystathionin beta synthase, selenium-binding protein 1, mercaptopyruvate sulphurtransferase, and thiosulphate sulphurtransferase. Higher mRNA expression of nuclear factor, erythroid 2-like 2, the main oxidative stress transcriptional factor in caecum, further supported our observations.
Collapse
|
50
|
Galectins in Endothelial Cell Biology and Angiogenesis: The Basics. Biomolecules 2021; 11:biom11091386. [PMID: 34572599 PMCID: PMC8464943 DOI: 10.3390/biom11091386] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 01/22/2023] Open
Abstract
Angiogenesis, the growth of new blood vessels out of existing vessels, is a complex and tightly regulated process. It is executed by the cells that cover the inner surface of the vasculature, i.e., the endothelial cells. During angiogenesis, these cells adopt different phenotypes, which allows them to proliferate and migrate, and to form tube-like structures that eventually result in the generation of a functional neovasculature. Multiple internal and external cues control these processes and the galectin protein family was found to be indispensable for proper execution of angiogenesis. Over the last three decades, several members of this glycan-binding protein family have been linked to endothelial cell functioning and to different steps of the angiogenesis cascade. This review provides a basic overview of our current knowledge regarding galectins in angiogenesis. It covers the main findings with regard to the endothelial expression of galectins and highlights their role in endothelial cell function and biology.
Collapse
|