1
|
Shil RK, Mohammed NBB, Dimitroff CJ. Galectin-9 - ligand axis: an emerging therapeutic target for multiple myeloma. Front Immunol 2024; 15:1469794. [PMID: 39386209 PMCID: PMC11461229 DOI: 10.3389/fimmu.2024.1469794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Galectin-9 (Gal-9) is a tandem-repeat galectin with diverse roles in immune homeostasis, inflammation, malignancy, and autoimmune diseases. In cancer, Gal-9 displays variable expression patterns across different tumor types. Its interactions with multiple binding partners, both intracellularly and extracellularly, influence key cellular processes, including immune cell modulation and tumor microenvironment dynamics. Notably, Gal-9 binding to cell-specific glycoconjugate ligands has been implicated in both promoting and suppressing tumor progression. Here, we provide insights into Gal-9 and its involvement in immune homeostasis and cancer biology with an emphasis on multiple myeloma (MM) pathophysiology, highlighting its complex and context-dependent dual functions as a pro- and anti-tumorigenic molecule and its potential implications for therapy in MM patients.
Collapse
Affiliation(s)
- Rajib K. Shil
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Norhan B. B. Mohammed
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Charles J. Dimitroff
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| |
Collapse
|
2
|
Meira C, Silva J, Quadros H, Silva L, Barreto B, Rocha V, Bomfim L, Santos E, Soares M. Galectins in Protozoan Parasitic Diseases: Potential Applications in Diagnostics and Therapeutics. Cells 2023; 12:2671. [PMID: 38067100 PMCID: PMC10705098 DOI: 10.3390/cells12232671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Neglected tropical diseases (NTDs) constitute a group of diseases that generally develop in tropical or subtropical climatic conditions and are related to poverty. Within the spectrum of NTDs, diseases caused by protozoa such as malaria, Chagas disease, and leishmaniasis exhibit elevated mortality rates, thereby constituting a substantial public health concern. Beyond their protozoan etiology, these NTDs share other similarities, such as the challenge of control and the lack of affordable, safe, and effective drugs. In view of the above, the need to explore novel diagnostic predictors and therapeutic targets for the treatment of these parasitic diseases is evident. In this context, galectins are attractive because they are a set of lectins bound to β-galactosides that play key roles in a variety of cellular processes, including host-parasite interaction such as adhesion and entry of parasites into the host cells, and participate in antiparasitic immunity in either a stimulatory or inhibitory manner, especially the galectins-1, -2, -3, and -9. These functions bestow upon galectins significant therapeutic prospects in the context of managing and diagnosing NTDs. Thus, the present review aims to elucidate the potential role of galectins in the diagnosis and treatment of malaria, leishmaniasis, and Chagas disease.
Collapse
Affiliation(s)
- Cássio Meira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 21040-900, Bahia, Brazil; (J.S.); (H.Q.); (L.S.); (B.B.); (V.R.); (L.B.)
- SENAI Institute of Innovation in Health Advanced Systems (ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Bahia, Brazil;
| | - Jaqueline Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 21040-900, Bahia, Brazil; (J.S.); (H.Q.); (L.S.); (B.B.); (V.R.); (L.B.)
| | - Helenita Quadros
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 21040-900, Bahia, Brazil; (J.S.); (H.Q.); (L.S.); (B.B.); (V.R.); (L.B.)
| | - Laís Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 21040-900, Bahia, Brazil; (J.S.); (H.Q.); (L.S.); (B.B.); (V.R.); (L.B.)
| | - Breno Barreto
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 21040-900, Bahia, Brazil; (J.S.); (H.Q.); (L.S.); (B.B.); (V.R.); (L.B.)
- SENAI Institute of Innovation in Health Advanced Systems (ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Bahia, Brazil;
- Institute of Health Sciences, Federal University of Bahia (UFBA), Salvador 40170-110, Bahia, Brazil
| | - Vinícius Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 21040-900, Bahia, Brazil; (J.S.); (H.Q.); (L.S.); (B.B.); (V.R.); (L.B.)
- SENAI Institute of Innovation in Health Advanced Systems (ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Bahia, Brazil;
| | - Larissa Bomfim
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 21040-900, Bahia, Brazil; (J.S.); (H.Q.); (L.S.); (B.B.); (V.R.); (L.B.)
| | - Emanuelle Santos
- SENAI Institute of Innovation in Health Advanced Systems (ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Bahia, Brazil;
| | - Milena Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 21040-900, Bahia, Brazil; (J.S.); (H.Q.); (L.S.); (B.B.); (V.R.); (L.B.)
- SENAI Institute of Innovation in Health Advanced Systems (ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Bahia, Brazil;
| |
Collapse
|
3
|
He R, Zhang Q, Xu L, Guo M, Gu X, Xie Y, Xu J, Shen Z. Characterization of a novel galectin in Sarcoptes scabiei and its role in regulating macrophage functions. Front Microbiol 2023; 14:1251475. [PMID: 37692380 PMCID: PMC10484005 DOI: 10.3389/fmicb.2023.1251475] [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: 07/07/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023] Open
Abstract
Sarcoptes scabiei (S. scabiei) endangers human and other mammalian health. There has been limited research into S. scabiei pathogenic mechanisms and the immunological interaction between S. scabiei and hosts. Galectins have critical roles in biological processes such as cell adhesion, signal transduction, and immune response mediation. Galectins of S. scabiei (SsGalectins) were cloned, expressed, and identified, and their transcriptional levels in S. scabiei were measured at various developmental stages. Fluorescent tissue localization was performed on SsGalectins of S. scabiei and scabies skin. A mouse AD model was constructed to evaluate the effect of rSsGalectins on skin pathogenic changes. Quantitative polymerase chain reaction and enzyme-linked immunoassay were used to identify macrophage polarization-related components and investigate the immunoregulatory effect of rSsGalectins on mouse macrophages. The results demonstrated that the S. scabiei infection causes macrophage infiltration in the scabies skin. The rSsGalectins displayed strong reactogenicity, and distinct genes of the SsGalectins were differently expressed in different developmental stages of S. scabiei. Fluorescence tissue localization revealed that the SsGalectins were mainly in the mouthparts, intestines, and body surface. Additionally, S. scabiei could secrete SsGalectins into the infected skin, proving that SsGalectins were excretion and secretion proteins of S. scabiei. In the mouse atopic dermatitis model, cutaneous macrophage infiltration and inflammation increase after rSsGalectins injection. Simultaneously, when rSsGalectins acted on bone marrow-derived macrophages, M1 macrophage-related polarization factors IL-1β, IL-6, and inducible nitric oxide synthase all increased, demonstrating that rSsGalectins can induce M1 polarization and produce pro-inflammatory cytokines. In conclusion, the SsGalectins are involved in the pathogenic process of S. scabiei by regulating the polarization of host macrophages to the M1 type when S. scabiei invade the host and promoting the incidence and development of the host's inflammatory response. This study offers fresh light on the pathogenic process of scabies mites, investigates the immunological interaction mechanism between S. scabiei and the host, and offers new insights into S. scabiei prevention and therapy.
Collapse
Affiliation(s)
- Ran He
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qian Zhang
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Luyang Xu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Maochuan Guo
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaobin Gu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yue Xie
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jing Xu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhaoli Shen
- College of Science, Sichuan Agricultural University, Ya'an, China
| |
Collapse
|
4
|
Morrison HM, Craft J, Rivera-Lugo R, Johnson JR, Golovkine GR, Bell SL, Dodd CE, Van Dis E, Beatty WL, Margolis SR, Repasy T, Shaker I, Lee AY, Vance RE, Stanley SA, Watson RO, Krogan NJ, Portnoy DA, Penn BH, Cox JS. Deficiency in Galectin-3, -8, and -9 impairs immunity to chronic Mycobacterium tuberculosis infection but not acute infection with multiple intracellular pathogens. PLoS Pathog 2023; 19:e1011088. [PMID: 37352334 PMCID: PMC10325092 DOI: 10.1371/journal.ppat.1011088] [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: 12/28/2022] [Revised: 07/06/2023] [Accepted: 05/01/2023] [Indexed: 06/25/2023] Open
Abstract
Macrophages employ an array of pattern recognition receptors to detect and eliminate intracellular pathogens that access the cytosol. The cytosolic carbohydrate sensors Galectin-3, -8, and -9 (Gal-3, Gal-8, and Gal-9) recognize damaged pathogen-containing phagosomes, and Gal-3 and Gal-8 are reported to restrict bacterial growth via autophagy in cultured cells. However, the contribution of these galectins to host resistance during bacterial infection in vivo remains unclear. We found that Gal-9 binds directly to Mycobacterium tuberculosis (Mtb) and Salmonella enterica serovar Typhimurium (Stm) and localizes to Mtb in macrophages. To determine the combined contribution of membrane damage-sensing galectins to immunity, we generated Gal-3, -8, and -9 triple knockout (TKO) mice. Mtb infection of primary macrophages from TKO mice resulted in defective autophagic flux but normal bacterial replication. Surprisingly, these mice had no discernable defect in resistance to acute infection with Mtb, Stm or Listeria monocytogenes, and had only modest impairments in bacterial growth restriction and CD4 T cell activation during chronic Mtb infection. Collectively, these findings indicate that while Gal-3, -8, and -9 respond to an array of intracellular pathogens, together these membrane damage-sensing galectins play a limited role in host resistance to bacterial infection.
Collapse
Affiliation(s)
- Huntly M. Morrison
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Julia Craft
- Department of Internal Medicine, Division of Infectious Diseases, University of California, Davis, Davis, California, United States of America
| | - Rafael Rivera-Lugo
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Jeffery R. Johnson
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco; Quantitative Biosciences Institute (QBI), University of California, San Francisco; Gladstone Institutes, San Francisco, California, United States of America
| | - Guillaume R. Golovkine
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Samantha L. Bell
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, School of Medicine, Bryan, Texas, United States of America
| | - Claire E. Dodd
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Erik Van Dis
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Wandy L. Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Shally R. Margolis
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Teresa Repasy
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Isaac Shaker
- Department of Internal Medicine, Division of Infectious Diseases, University of California, Davis, Davis, California, United States of America
| | - Angus Y. Lee
- Cancer Research Laboratory, University of California, Berkeley, Berkeley, California, United States of America
| | - Russell E. Vance
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - Sarah A. Stanley
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
- School of Public Health, Division of Infectious Diseases and Vaccinology, University of California, Berkeley, Berkeley, California, United States of America
| | - Robert O. Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health, School of Medicine, Bryan, Texas, United States of America
| | - Nevan J. Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco; Quantitative Biosciences Institute (QBI), University of California, San Francisco; Gladstone Institutes, San Francisco, California, United States of America
| | - Daniel A. Portnoy
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| | - Bennett H. Penn
- Department of Internal Medicine, Division of Infectious Diseases, University of California, Davis, Davis, California, United States of America
| | - Jeffery S. Cox
- Department of Molecular and Cell Biology, Division of Immunology and Molecular Medicine, University of California, Berkeley, Berkeley, California, United States of America
| |
Collapse
|
5
|
Abstract
Leishmaniasis is a zoonotic and vector-borne infectious disease that is caused by the genus Leishmania belonging to the trypanosomatid family. The protozoan parasite has a digenetic life cycle involving a mammalian host and an insect vector. Leishmaniasisis is a worldwide public health problem falling under the neglected tropical disease category, with over 90 endemic countries, and approximately 1 million new cases and 20,000 deaths annually. Leishmania infection can progress toward the development of species–specific pathologic disorders, ranging in severity from self-healing cutaneous lesions to disseminating muco-cutaneous and fatal visceral manifestations. The severity and the outcome of leishmaniasis is determined by the parasite’s antigenic epitope characteristics, the vector physiology, and most importantly, the immune response and immune status of the host. This review examines the nature of host–pathogen interaction in leishmaniasis, innate and adaptive immune responses, and various strategies that have been employed for vaccine development.
Collapse
|
6
|
Taslimi Y, Masoudzadeh N, Bahrami F, Rafati S. Cutaneous leishmaniasis: multiomics approaches to unravel the role of immune cells checkpoints. Expert Rev Proteomics 2022; 19:213-225. [PMID: 36191333 DOI: 10.1080/14789450.2022.2131545] [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: 01/07/2023]
Abstract
INTRODUCTION Cutaneous leishmaniasis (CL) is the most frequent form of leishmaniases, associated with skin inflammation and ulceration. Understanding the interaction of different phagocytic cells in the recognition and uptake of different Leishmania species is critical for controlling the infection. Phagocytic cells have a pivotal role as professional antigen-presenting cells that bridge the innate and adaptive immunity and shape the outcome of the disease. AREAS COVERED Here we reviewed new technologies with high-throughput data collection capabilities along with systems biology approaches which are recently being used to decode the paradox of CL immunology. EXPERT OPINION We emphasized on the crosstalk between DC and T-cells while focusing on the immune checkpoints interactions between the human immune system and the Leishmania species. Further, we discussed omics technologies including bulk RNA sequencing, reverse transcriptase-multiplex ligation dependent probe amplification (RT-MLPA), and proximity extension assay (PEA) in studies on human blood or tissue-driven samples from CL patients in which we have so far been involved.
Collapse
Affiliation(s)
- Yasaman Taslimi
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran Iran
| | - Nasrin Masoudzadeh
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran Iran
| | - Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran Iran
| |
Collapse
|
7
|
Maruszewska-Cheruiyot M, Stear M, Donskow-Łysoniewska K. Galectins - Important players of the immune response to CNS parasitic infection. Brain Behav Immun Health 2021; 13:100221. [PMID: 34589740 PMCID: PMC8474370 DOI: 10.1016/j.bbih.2021.100221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/18/2021] [Accepted: 01/30/2021] [Indexed: 11/18/2022] Open
Abstract
Galectins are a family of proteins that bind β-galactosides and play key roles in a variety of cellular processes including host defense and entry of parasites into the host cells. They have been well studied in hosts but less so in parasites. As both host and parasite galectins are highly upregulated proteins following infection, galectins are an area of increasing interest and their role in immune modulation has only recently become clear. Correlation of CNS parasitic diseases with mental disorders as a result of direct or indirect interaction has been observed. Therefore, galectins produced by the parasite should be taken into consideration as potential therapeutic agents.
Collapse
Affiliation(s)
- Marta Maruszewska-Cheruiyot
- Laboratory of Parasitology, General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163, Warsaw, Poland
- Corresponding author.
| | - Michael Stear
- Department of Animal, Plant and Soil Science, Agribio, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Katarzyna Donskow-Łysoniewska
- Laboratory of Parasitology, General Karol Kaczkowski Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163, Warsaw, Poland
| |
Collapse
|
8
|
Design and characterization of high-affinity synthetic peptides as bioreceptors for diagnosis of cutaneous leishmaniasis. Anal Bioanal Chem 2021; 413:4545-4555. [PMID: 34037808 PMCID: PMC8149292 DOI: 10.1007/s00216-021-03424-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/12/2021] [Accepted: 05/20/2021] [Indexed: 11/01/2022]
Abstract
Cutaneous leishmaniasis (CL) is one of the illnesses caused by Leishmania parasite infection, which can be asymptomatic or severe according to the infecting Leishmania strain. CL is commonly diagnosed by directly detecting the parasites or their DNA in tissue samples. New diagnostic methodologies target specific proteins (biomarkers) secreted by the parasite during the infection process. However, specific bioreceptors for the in vivo or in vitro detection of these novel biomarkers are rather limited in terms of sensitivity and specificity. For this reason, we here introduce three novel peptides as bioreceptors for the highly sensitive and selective identification of acid phosphatase (sAP) and proteophosphoglycan (PPG), which have a crucial role in leishmaniasis infection. These high-affinity peptides have been designed from the conservative domains of the lectin family, holding the ability to interact with the biological target and produce the same effect than the original protein. The synthetic peptides have been characterized and the affinity and kinetic constants for their interaction with the targets (sAP and PPG) have been determined by a surface plasmon resonance biosensor. Values obtained for KD are in the nanomolar range, which is comparable to high-affinity antibodies, with the additional advantage of a high biochemical stability and simpler production. Pep2854 exhibited a high affinity for sAP (KD = 1.48 nM) while Pep2856 had a good affinity for PPG (KD 1.76 nM). This study evidences that these peptidomimetics represent a novel alternative tool to the use of high molecular weight proteins for biorecognition in the diagnostic test and biosensor devices for CL.
Collapse
|
9
|
Pfeifer L, Baumann A, Petersen LM, Höger B, Beitz E, Classen B. Degraded Arabinogalactans and Their Binding Properties to Cancer-Associated Human Galectins. Int J Mol Sci 2021; 22:ijms22084058. [PMID: 33920014 PMCID: PMC8071012 DOI: 10.3390/ijms22084058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022] Open
Abstract
Galectins represent β-galactoside-binding proteins with numerous functions. Due to their role in tumor progression, human galectins-1, -3 and -7 (Gal-1, -3 and -7) are potential targets for cancer therapy. As plant derived glycans might act as galectin inhibitors, we prepared galactans by partial degradation of plant arabinogalactan-proteins. Besides commercially purchased galectins, we produced Gal-1 and -7 in a cell free system and tested binding capacities of the galectins to the galactans by biolayer-interferometry. Results for commercial and cell-free expressed galectins were comparable confirming functionality of the cell-free produced galectins. Our results revealed that galactans from Echinacea purpurea bind to Gal-1 and -7 with KD values of 1–2 µM and to Gal-3 slightly stronger with KD values between 0.36 and 0.70 µM depending on the sensor type. Galactans from the seagrass Zostera marina with higher branching of the galactan and higher content of uronic acids showed stronger binding to Gal-3 (0.08–0.28 µM) compared to galactan from Echinacea. The results contribute to knowledge on interactions between plant polysaccharides and galectins. Arabinogalactan-proteins have been identified as a new source for production of galactans with possible capability to act as galectin inhibitors.
Collapse
Affiliation(s)
- Lukas Pfeifer
- Department of Pharmaceutical Biology, Pharmaceutical Institute, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany; (L.P.); (A.B.)
| | - Alexander Baumann
- Department of Pharmaceutical Biology, Pharmaceutical Institute, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany; (L.P.); (A.B.)
| | - Lea Madlen Petersen
- Department of Pharmaceutical Chemistry, Pharmaceutical Institute, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany; (L.M.P.); (B.H.); (E.B.)
| | - Bastian Höger
- Department of Pharmaceutical Chemistry, Pharmaceutical Institute, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany; (L.M.P.); (B.H.); (E.B.)
| | - Eric Beitz
- Department of Pharmaceutical Chemistry, Pharmaceutical Institute, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany; (L.M.P.); (B.H.); (E.B.)
| | - Birgit Classen
- Department of Pharmaceutical Biology, Pharmaceutical Institute, Christian-Albrechts-University of Kiel, 24118 Kiel, Germany; (L.P.); (A.B.)
- Correspondence: ; Tel.: +49-431-8801130
| |
Collapse
|
10
|
Nunoue T, Yamaguchi S, Teshigawara S, Katayama A, Nakatsuka A, Eguchi J, Niki T, Wada J. Lgals9 deficiency ameliorates obesity by modulating redox state of PRDX2. Sci Rep 2021; 11:5991. [PMID: 33727589 PMCID: PMC7966757 DOI: 10.1038/s41598-021-85080-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/25/2021] [Indexed: 01/25/2023] Open
Abstract
The adipose tissue is regarded as an endocrine organ and secretes bioactive adipokines modulating chronic inflammation and oxidative stress in obesity. Gal-9 is secreted out upon cell injuries, interacts with T-cell immunoglobulin-3 (Tim-3) and induces apoptosis in activated Th1 cells. Gal-9 also binds to protein disulfide isomerase (PDI), maintains PDI on surface of T cells, and increases free thiols in the disulfide/thiol cycles. To explore the molecular mechanism of obesity, we investigated Gal-9−/− and Gal-9wt/wt C57BL/6J mice fed with high fat-high sucrose (HFHS) chow. Gal-9−/− mice were resistant to diet-induced obesity associated with reduction of epididymal and mesenteric fat tissues and improved glucose tolerance compared with Gal-9wt/wt mice. However, the number of M1, M2 macrophages, and M1/M2 ratio in epididymal fat were unaltered. Under HFHS chow, Gal-9−/− mice receiving Gal-9−/− or Gal-9wt/wt bone marrow-derived cells (BMCs) demonstrated significantly lower body weight compared with Gal-9wt/wt mice receiving Gal-9−/− BMCs. We identified the binding between Gal-9 and peroxiredoxin-2 (PRDX2) in sugar chain-independent manner by nanoLC-MS/MS, immunoprecipitation, and pull-down assay. In 3T3L1 adipocytes, Gal-9 knockdown shifts PRDX2 monomer (reduced form) dominant from PRDX2 dimer (oxidized form) under oxidative stress with H2O2. The inhibition of Gal-9 in adipocytes may be a new therapeutic approach targeting the oxidative stress and subsequent glucose intolerance in obesity.
Collapse
Affiliation(s)
- Tomokazu Nunoue
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Satoshi Yamaguchi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Sanae Teshigawara
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Akihiro Katayama
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Atsuko Nakatsuka
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Jun Eguchi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Toshiro Niki
- Department of Immunology, Kagawa University, Takamatsu, Kagawa, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| |
Collapse
|
11
|
Moar P, Tandon R. Galectin-9 as a biomarker of disease severity. Cell Immunol 2021; 361:104287. [PMID: 33494007 DOI: 10.1016/j.cellimm.2021.104287] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/26/2020] [Accepted: 01/09/2021] [Indexed: 12/16/2022]
Abstract
Galectin-9 (Gal-9) is a β-galactoside binding lectin known for its immunomodulatory role in various microbial infections. Gal-9 is expressed in all organ systems and localized in the nucleus, cell surface, cytoplasm and the extracellular matrix. It mediates host-pathogen interactions and regulates cell signalling via binding to its receptors. Gal-9 is involved in many physiological functions such as cell growth, differentiation, adhesion, communication and death. However, recent studies have emphasized on the elevated levels of Gal-9 in autoimmune disorders, viral infections, parasitic invasion, cancer, acute liver failure, atopic dermatitis, chronic kidney disease, type-2 diabetes, coronary artery disease, atherosclerosis and benign infertility-related gynecological disorders. In this paper we have reviewed the potential of Gal-9 as a reliable, sensitive and non-invasive biomarker of disease severity. Tracking changes in Gal-9 levels and its implementation as a biomarker in clinical practice will be an important tool to monitor disease activity and facilitate personalized treatment decisions.
Collapse
Affiliation(s)
- Preeti Moar
- Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| | - Ravi Tandon
- Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
12
|
dos Santos Meira C, Gedamu L. Protective or Detrimental? Understanding the Role of Host Immunity in Leishmaniasis. Microorganisms 2019; 7:microorganisms7120695. [PMID: 31847221 PMCID: PMC6956275 DOI: 10.3390/microorganisms7120695] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/04/2019] [Accepted: 12/10/2019] [Indexed: 02/06/2023] Open
Abstract
The intracellular protozoan parasites of the genus Leishmania are the causative agents of leishmaniasis, a vector-borne disease of major public health concern, estimated to affect 12 million people worldwide. The clinical manifestations of leishmaniasis are highly variable and can range from self-healing localized cutaneous lesions to life-threatening disseminated visceral disease. Once introduced into the skin by infected sandflies, Leishmania parasites interact with a variety of immune cells, such as neutrophils, monocytes, dendritic cells (DCs), and macrophages. The resolution of infection requires a finely tuned interplay between innate and adaptive immune cells, culminating with the activation of microbicidal functions and parasite clearance within host cells. However, several factors derived from the host, insect vector, and Leishmania spp., including the presence of a double-stranded RNA virus (LRV), can modulate the host immunity and influence the disease outcome. In this review, we discuss the immune mechanisms underlying the main forms of leishmaniasis, some of the factors involved with the establishment of infection and disease severity, and potential approaches for vaccine and drug development focused on host immunity.
Collapse
|
13
|
Pineda M, Corvo L, Callejas-Hernández F, Fresno M, Bonay P. Trypanosoma cruzi cleaves galectin-3 N-terminal domain to suppress its innate microbicidal activity. Clin Exp Immunol 2019; 199:216-229. [PMID: 31593356 DOI: 10.1111/cei.13379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2019] [Indexed: 12/01/2022] Open
Abstract
Galectin-3 is the best-characterized member of galectins, an evolutionary conserved family of galactoside-binding proteins that play central roles in infection and immunity, regulating inflammation, cell migration and cell apoptosis. Differentially expressed by cells and tissues with immune privilege, they bind not only to host ligands, but also to glycans expressed by pathogens. In this regard, we have previously shown that human galectin-3 recognizes several genetic lineages of the protozoan parasite Trypanosoma cruzi, the causal agent of Chagas' disease or American trypanosomiasis. Herein we describe a molecular mechanism developed by T. cruzi to proteolytically process galectin-3 that generates a truncated form of the protein lacking its N-terminal domain - required for protein oligomerization - but still conserves a functional carbohydrate recognition domain (CRD). Such processing relies on specific T. cruzi proteases, including Zn-metalloproteases and collagenases, and ultimately conveys profound changes in galectin-3-dependent effects, as chemical inhibition of parasite proteases allows galectin-3 to induce parasite death in vitro. Thus, T. cruzi might have established distinct mechanisms to counteract galectin-3-mediated immunity and microbicide properties. Interestingly, non-pathogenic T. rangeli lacked the ability to cleave galectin-3, suggesting that during evolution two genetically similar organisms have developed different molecular mechanisms that, in the case of T. cruzi, favoured its pathogenicity, highlighting the importance of T. cruzi proteases to avoid immune mechanisms triggered by galectin-3 upon infection. This study provides the first evidence of a novel strategy developed by T. cruzi to abrogate signalling mechanisms associated with galectin-3-dependent innate immunity.
Collapse
Affiliation(s)
- M Pineda
- Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Nicolás Cabrera 1, Madrid, Spain
| | - L Corvo
- Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Nicolás Cabrera 1, Madrid, Spain
| | - F Callejas-Hernández
- Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Nicolás Cabrera 1, Madrid, Spain
| | - M Fresno
- Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Nicolás Cabrera 1, Madrid, Spain
| | - P Bonay
- Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Nicolás Cabrera 1, Madrid, Spain
| |
Collapse
|
14
|
CRISPR/Cas9 Mutagenesis in Phlebotomus papatasi: the Immune Deficiency Pathway Impacts Vector Competence for Leishmania major. mBio 2019; 10:mBio.01941-19. [PMID: 31455654 PMCID: PMC6712399 DOI: 10.1128/mbio.01941-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sand flies are the natural vectors for the Leishmania species that produce a spectrum of diseases in their mammalian hosts, including humans. Studies of sand fly/Leishmania interactions have been limited by the absence of genome editing techniques applicable to these insects. In this report, we adapted CRISPR (clustered regularly interspaced palindromic repeat)/Cas9 (CRISPR-associated protein 9) technology to the Phlebotomus papatasi sand fly, a natural vector for Leishmania major, targeting the sand fly immune deficiency (IMD) pathway in order to decipher its contribution to vector competence. We established a protocol for transformation in P. papatasi and were able to generate transmissible null mutant alleles for Relish (Rel), the only transcription factor of the IMD pathway. While the maintenance of a homozygous mutant stock was severely compromised, we were able to establish in an early generation their greater susceptibility to infection with L. major Flies carrying different heterozygous mutant alleles variably displayed a more permissive phenotype, presenting higher loads of parasites or greater numbers of infective-stage promastigotes. Together, our data show (i) the successful adaptation of the CRISPR/Cas9 technology to sand flies and (ii) the impact of the sand fly immune response on vector competence for Leishmania parasites.IMPORTANCE Sand flies are the natural vectors of Leishmania parasites. Studies of sand fly/Leishmania interactions have been limited by the lack of successful genomic manipulation of these insects. This paper shows the first example of successful targeted mutagenesis in sand flies via adaptation of the CRISPR/Cas9 editing technique. We generated transmissible null mutant alleles of relish, a gene known to be essential for the control of immune response in other insects. In addition to the expected higher level of susceptibility to bacteria, the mutant flies presented higher loads of parasites when infected with L. major, showing that the sand fly immune response impacts its vector competence for this pathogen.
Collapse
|
15
|
Sato S. Cytosolic Galectins and Their Release and Roles as Carbohydrate-Binding Proteins in Host–Pathogen Interaction. TRENDS GLYCOSCI GLYC 2018. [DOI: 10.4052/tigg.1739.1se] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Sachiko Sato
- Glycobiology and Bioimaging Laboratory, Research Centre for Infectious Diseases, Faculty of Medicine, Laval University
| |
Collapse
|
16
|
Iwaki J, Hirabayashi J. Carbohydrate-Binding Specificity of Human Galectins: An Overview by Frontal Affinity Chromatography. TRENDS GLYCOSCI GLYC 2018. [DOI: 10.4052/tigg.1728.1se] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Jun Iwaki
- National Institute of Advanced Industrial Science and Technology
| | - Jun Hirabayashi
- National Institute of Advanced Industrial Science and Technology
| |
Collapse
|
17
|
The roles of galectins in parasitic infections. Acta Trop 2018; 177:97-104. [PMID: 28986248 DOI: 10.1016/j.actatropica.2017.09.027] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/10/2017] [Accepted: 09/29/2017] [Indexed: 12/18/2022]
Abstract
Galectins is a family of multifunctional lectins. Fifteen galectins have been identified from a variety of cells and tissues of vertebrates and invertebrates. Galectins have been shown to play pivotal roles in host-pathogen interaction such as adhesion of pathogens to host cells and activation of host innate and adaptive immunity. In recent years, the roles of galectins during parasite infections have gained increasing attention. Galectins produced by different hosts can act as pattern recognition receptors detecting conserved pathogen-associated molecular patterns of parasites, while galectins produced by parasites can modulate host responses. This review summarizes some recent studies on the roles of galectins produced by parasitic protozoa, nematodes, and trematodes and their hosts. Understanding the roles of galectins in host-parasite interactions may provide targets for immune intervention and therapies of parasitic infections.
Collapse
|
18
|
Sun Y, Li Y, Wu Y, Xiong L, Li C, Wang C, Li D, Lan J, Zhang Z, Jing B, Gu X, Xie Y, Lai W, Peng X, Yang G. Fatty-binding protein and galectin of Baylisascaris schroederi: Prokaryotic expression and preliminary evaluation of serodiagnostic potential. PLoS One 2017; 12:e0182094. [PMID: 28750056 PMCID: PMC5531546 DOI: 10.1371/journal.pone.0182094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/12/2017] [Indexed: 11/18/2022] Open
Abstract
Baylisascaris schroederi is a common parasite of captive giant pandas. The diagnosis of this ascariasis is normally carried out by a sedimentation-floatation method or PCR to detect eggs in feces, but neither method is suitable for early diagnosis. Fatty acid-binding protein (FABP) and galectin (GAL) exist in various animals and participate in important biology of parasites. Because of their good immunogenicity, they are seen as potential antigens for the diagnosis of parasitic diseases. In this study, we cloned and expressed recombinant FABP and GAL from B. schroederi (rBs-FABP and rBs-GAL) and developed indirect enzyme-linked immunosorbent assays (ELISAs) to evaluate their potential for diagnosing ascariasis in giant pandas. Immunolocalization showed that Bs-FABP and Bs-GAL were widely distributed in adult worms. The ELISA based on rBs-FABP showed sensitivity of 95.8% (23/24) and specificity of 100% (12/12), and that based on rBs-GAL had sensitivity of 91.7% (22/24) and specificity of 100% (12/12).
Collapse
Affiliation(s)
- Ying Sun
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yu Li
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yiran Wu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lang Xiong
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Caiwu Li
- China Conservation and Research Center for Giant Panda, Wolong, China
| | - Chengdong Wang
- China Conservation and Research Center for Giant Panda, Wolong, China
| | - Desheng Li
- China Conservation and Research Center for Giant Panda, Wolong, China
| | - Jingchao Lan
- Chengdu Research Base of Giant Panda Breeding, Chengdu, China
| | - Zhihe Zhang
- Chengdu Research Base of Giant Panda Breeding, Chengdu, China
| | - Bo Jing
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaobing Gu
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yue Xie
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Weimin Lai
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xuerong Peng
- College of Science, Sichuan Agricultural University, Ya’an, China
| | - Guangyou Yang
- Department of Parasitology, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- * E-mail:
| |
Collapse
|
19
|
Abstract
Galectins is a family of non-classically secreted, beta-galactoside-binding proteins that has recently received considerable attention in the spatio-temporal regulation of surface 'signal lattice' organization, membrane dynamics, cell-adhesion and disease therapeutics. Galectin-9 is a unique member of this family, with two non-homologous carbohydrate recognition domains joined by a linker peptide sequence of variable lengths, generating isoforms with distinct properties and functions in both physiological and pathological settings, such as during development, immune reaction, neoplastic transformations and metastasis. In this review, we summarize the latest knowledge on the structure, receptors, cellular targets, trafficking pathways and functional properties of galectin-9 and discuss how galectin-9-mediated signalling cascades can be exploited in cancers and immunotherapies.
Collapse
Affiliation(s)
- Sebastian John
- Department of Neurobiology and Genetics, Division of Disease Biology, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram 695014, India
| | | |
Collapse
|
20
|
Arthur CM, Patel SR, Mener A, Kamili NA, Fasano RM, Meyer E, Winkler AM, Sola-Visner M, Josephson CD, Stowell SR. Innate immunity against molecular mimicry: Examining galectin-mediated antimicrobial activity. Bioessays 2016; 37:1327-37. [PMID: 26577077 DOI: 10.1002/bies.201500055] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Adaptive immunity provides the unique ability to respond to a nearly infinite range of antigenic determinants. Given the inherent plasticity of the adaptive immune system, a series of tolerance mechanisms exist to reduce reactivity toward self. While this reduces the probability of autoimmunity, it also creates an important gap in adaptive immunity: the ability to recognize microbes that look like self. As a variety of microbes decorate themselves in self-like carbohydrate antigens and tolerance reduces the ability of adaptive immunity to react with self-like structures, protection against molecular mimicry likely resides within the innate arm of immunity. In this review, we will explore the potential consequences of microbial molecular mimicry, including factors within innate immunity that appear to specifically target microbes expressing self-like antigens, and therefore provide protection against molecular mimicry.
Collapse
Affiliation(s)
- Connie M Arthur
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA
| | - Seema R Patel
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA
| | - Amanda Mener
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA
| | - Nourine A Kamili
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA
| | - Ross M Fasano
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA
| | - Erin Meyer
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA
| | - Annie M Winkler
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA
| | - Martha Sola-Visner
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Cassandra D Josephson
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA
| | - Sean R Stowell
- Department of Laboratory Medicine and Pathology, Center for Transfusion Medicine and Cellular Therapies, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
21
|
Liu J, Huang S, Su XZ, Song J, Lu F. Blockage of Galectin-receptor Interactions by α-lactose Exacerbates Plasmodium berghei-induced Pulmonary Immunopathology. Sci Rep 2016; 6:32024. [PMID: 27554340 PMCID: PMC4995515 DOI: 10.1038/srep32024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 08/02/2016] [Indexed: 12/22/2022] Open
Abstract
Malaria-associated acute lung injury (ALI) is a frequent complication of severe malaria that is often caused by "excessive" immune responses. To better understand the mechanism of ALI in malaria infection, here we investigated the roles of galectin (Gal)-1, 3, 8, 9 and the receptors of Gal-9 (Tim-3, CD44, CD137, and PDI) in malaria-induced ALI. We injected alpha (α)-lactose into mice-infected with Plasmodium berghei ANKA (PbANKA) to block galectins and found significantly elevated total proteins in bronchoalveolar lavage fluid, higher parasitemia and tissue parasite burden, and increased numbers of CD68(+) alveolar macrophages as well as apoptotic cells in the lungs after blockage. Additionally, mRNA levels of Gal-9, Tim-3, CD44, CD137, and PDI were significantly increased in the lungs at day 5 after infection, and the levels of CD137, IFN-α, IFN-β, IFN-γ, IL-4, and IL-10 in the lungs were also increased after α-lactose treatment. Similarly, the levels of Gal-9, Tim-3, IFN-α, IFN-β, IFN-γ, and IL-10 were all significantly increased in murine peritoneal macrophages co-cultured with PbANKA-infected red blood cells in vitro; but only IFN-α and IFN-β were significantly increased after α-lactose treatment. Our data indicate that Gal-9 interaction with its multiple receptors play an important role in murine malaria-associated ALI.
Collapse
Affiliation(s)
- Jinfeng Liu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, Guangdong, China
| | - Shiguang Huang
- School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China
| | - Xin-Zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States of America.,State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, China
| | - Jianping Song
- Institute of Science and Technology, Guangzhou University of Chinese Medicine, 436 Chentai Road, Baiyun District, Guangzhou 510445, Guangdong, China
| | - Fangli Lu
- Department of Parasitology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China.,Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, Guangdong, China
| |
Collapse
|
22
|
Dembele BPP, Chagan-Yasutan H, Niki T, Ashino Y, Tangpukdee N, Shinichi E, Krudsood S, Kano S, Hattori T. Plasma levels of Galectin-9 reflect disease severity in malaria infection. Malar J 2016; 15:403. [PMID: 27515948 PMCID: PMC4982308 DOI: 10.1186/s12936-016-1471-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/02/2016] [Indexed: 12/17/2022] Open
Abstract
Background Galectin-9 (Gal-9) is a β-galactoside-binding lectin that interacts with sugar moieties on glycoproteins and glycolipids of cells and pathogens. Gal-9 is known as an immune modulator that induces cell death via interaction with T cell immunoglobulin and mucin domain-3 (Tim3), a co-inhibitory receptor, and it inhibits production of several pro-inflammatory cytokines (TNF, IL-6 and IL-1α) and enhances production of IL-10. To understand the immune pathology of malaria, the Gal-9 in plasma was measured. Methods Plasma samples and clinical parameters were obtained from 50 acute malaria cases (nine severe and 41 uncomplicated cases) from Thailand at three time points: day 0, day 7 and day 28. Gal-9 levels were determined by ELISA. A total of 38 species of cytokines and chemokines were measured using a BioPlex assay. Results Gal-9 levels were higher at day 0 compared to day 7 and day 28 (P < 0.0001). Gal-9 levels were also higher in severe malaria (SM) cases compared to uncomplicated (UM) cases at day 0 and day 7 (923 vs 617 pg/mL; P = 0.03, and 659 vs 348 pg/mL; P = 0.02 respectively). Median Gal-9 levels were higher in patients with blood urea nitrogen to creatinine ratio (BUN/creatinine) ≥20 (mg/dL) than in patients with BUN/creatinine <20 (mg/dL) at day 0 (817.3 vs 576.2 pg/mL, P = 0.007). Gal-9 was inversely significantly correlated with chloride levels in both SM and UM cases (rs = −0.73 and rs = −0.46, respectively). In both UM and SM cases, Gal-9 was significantly associated with pro- and anti-inflammatory cytokines and chemokines such as TNF, IL-6, IFN-α2, IFN-γ, IL-1Ra and IL-10. These correlations were observed at day 0 but disappeared at day 28. Conclusions Gal-9 is released during acute malaria, and reflects its severity. This elevation of Gal-9 in acute malaria infection raises the possibility of its role in termination of the immune response by binding to Tim-3, a receptor of Gal-9.
Collapse
Affiliation(s)
- Bindongo P P Dembele
- Division of International Cooperation for Disaster Medicine, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Haorile Chagan-Yasutan
- Division of Disaster-related Infectious Disease, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan.,Emerging Infectious Diseases, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Toshiro Niki
- Department of Immunology, Kagawa University, Takamatsu, Japan.,GalPharma Co., Ltd., Takamatsu, Japan
| | - Yugo Ashino
- Division of Disaster-related Infectious Disease, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan.,Emerging Infectious Diseases, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Noppadon Tangpukdee
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Egawa Shinichi
- Division of International Cooperation for Disaster Medicine, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan
| | - Srivicha Krudsood
- Clinical Malaria Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Shigeyuki Kano
- Department of Tropical Medicine and Malaria, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Toshio Hattori
- Division of Disaster-related Infectious Disease, International Research Institute of Disaster Science, Tohoku University, Sendai, Japan. .,Emerging Infectious Diseases, Graduate School of Medicine, Tohoku University, Sendai, Japan. .,Department of Occupational Therapy, Graduate School of Health Science Studies, Kibi International University, 8 Igamachi, Takahashi, Okayama, Japan.
| |
Collapse
|
23
|
Li Y, Yuan C, Wang L, Lu M, Wang Y, Wen Y, Yan R, Xu L, Song X, Li X. Transmembrane protein 147 (TMEM147): another partner protein of Haemonchus contortus galectin on the goat peripheral blood mononuclear cells (PBMC). Parasit Vectors 2016; 9:355. [PMID: 27337943 PMCID: PMC4918192 DOI: 10.1186/s13071-016-1640-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/13/2016] [Indexed: 12/03/2022] Open
Abstract
Background Recombinant galectins of male and female Haemonchus contortus (rHco-gal-m/f) have been recognized as significant regulators of the functions of goat peripheral blood mononuclear cells (PBMC). In previous research, transmembrane protein 63A (TMEM63A) was identified as a partner protein in the regulation associated with H. contortus infection. However, in the identification of binding partners for galectins of male and female H. contortus (Hco-gal-m/f) by yeast two-hybrid (YTH) screening, it was found that the transmembrane protein 147 (TMEM147) could also bind to Hco-gal-m/f. In this study, the functions of TMEM147 in the regulations of H. contortus galectin on the goat PBMC were investigated. Methods To identify Hco-gal-m/f-interacting proteins, a yeast two-hybrid system to detect interactions was used. Co-immunoprecipitation and immunoblotting were used to validate the interaction between recombinant galectins of male H. contortus (rHco-gal-m) and candidate binding protein. The localization of TMEM147 in PBMC was explored by immunofluorescence in confocal imaging studies. Flow cytometry was used to determine the distribution of TMEM147 in T cells, B cells and monocytes in PBMC. The modulatory effects of rHco-gal-m and TMEM147 on cell proliferation, phagocytosis, nitric oxide production, migration, apoptosis and cytokine mRNA transcription were observed by co-incubation of rHco-gal-m and knockdown of the tmem147 gene. Results In this research, it was demonstrated that TMEM147 could bind to rHco-gal-m/f. Immunofluorescence assays showed that TMEM147 was localized to the cell membrane and within the cell membrane in goat PBMC. Flow cytometric analysis revealed that TMEM147 was expressed in all B cells and monocytes in goat PBMC. However, 3.8 % of T cells did not express this protein. Knockdown of the tmem147 gene using RNA interference (RNAi) showed that the interaction of galectin with TMEM147 mainly mediated cell proliferation, cell apoptosis, transcription of interleukin-10 (IL-10) and transforming growth factor-β1 (TGF-β1) of goat PBMC. This membrane protein, together with TMEM63A, was also related to the regulation of galectin on phagocytosis and nitric oxide production of goat PBMC. However, it might not be involved in the regulation of galectin on the migration and interferon-γ (IFN-γ) transcription of goat PBMC. Conclusions Our results showed that TMEM147 was a binding partner of Hco-gal-m/f and mediated the immunological regulation of Hco-gal-m/f on goat PBMC in a manner different to that of TMEM63A. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1640-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Cheng Yuan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - LiKun Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - MingMin Lu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - YuJian Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - YuLing Wen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - RuoFeng Yan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - LiXin Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - XiaoKai Song
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - XiangRui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| |
Collapse
|
24
|
Nita-Lazar M, Mancini J, Feng C, González-Montalbán N, Ravindran C, Jackson S, de Las Heras-Sánchez A, Giomarelli B, Ahmed H, Haslam SM, Wu G, Dell A, Ammayappan A, Vakharia VN, Vasta GR. The zebrafish galectins Drgal1-L2 and Drgal3-L1 bind in vitro to the infectious hematopoietic necrosis virus (IHNV) glycoprotein and reduce viral adhesion to fish epithelial cells. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 55:241-252. [PMID: 26429411 PMCID: PMC4684960 DOI: 10.1016/j.dci.2015.09.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 09/17/2015] [Accepted: 09/17/2015] [Indexed: 06/05/2023]
Abstract
The infectious hematopoietic necrosis virus (IHNV; Rhabdoviridae, Novirhabdovirus) infects teleost fish, such as salmon and trout, and is responsible for significant losses in the aquaculture industry and in wild fish populations. Although IHNV enters the host through the skin at the base of the fins, the viral adhesion and entry mechanisms are not fully understood. In recent years, evidence has accumulated in support of the key roles played by protein-carbohydrate interactions between host lectins secreted to the extracellular space and virion envelope glycoproteins in modulating viral adhesion and infectivity. In this study, we assessed in vitro the potential role(s) of zebrafish (Danio rerio) proto type galectin-1 (Drgal1-L2) and a chimera galectin-3 (Drgal3-L1) in IHNV adhesion to epithelial cells. Our results suggest that the extracellular Drgal1-L2 and Drgal3-L1 interact directly and in a carbohydrate-dependent manner with the IHNV glycosylated envelope and glycans on the epithelial cell surface, significantly reducing viral adhesion.
Collapse
Affiliation(s)
- Mihai Nita-Lazar
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, Maryland, USA
| | - Justin Mancini
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, Maryland, USA
| | - Chiguang Feng
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, Maryland, USA
| | - Núria González-Montalbán
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, Maryland, USA
| | - Chinnarajan Ravindran
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, Maryland, USA
| | - Shawn Jackson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, Maryland, USA
| | - Ana de Las Heras-Sánchez
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, Maryland, USA
| | - Barbara Giomarelli
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, Maryland, USA
| | - Hafiz Ahmed
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, Maryland, USA
| | - Stuart M Haslam
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College, London, UK
| | - Gang Wu
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College, London, UK
| | - Anne Dell
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College, London, UK
| | - Arun Ammayappan
- Department of Marine Biotechnology, University of Maryland Baltimore County, Baltimore, Maryland, USA
| | - Vikram N Vakharia
- Department of Marine Biotechnology, University of Maryland Baltimore County, Baltimore, Maryland, USA
| | - Gerardo R Vasta
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Institute of Marine and Environmental Technology, Baltimore, Maryland, USA
| |
Collapse
|
25
|
Fichorova RN, Yamamoto HS, Fashemi T, Foley E, Ryan S, Beatty N, Dawood H, Hayes GR, St-Pierre G, Sato S, Singh BN. Trichomonas vaginalis Lipophosphoglycan Exploits Binding to Galectin-1 and -3 to Modulate Epithelial Immunity. J Biol Chem 2016; 291:998-1013. [PMID: 26589797 PMCID: PMC4705417 DOI: 10.1074/jbc.m115.651497] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 10/29/2015] [Indexed: 11/06/2022] Open
Abstract
Trichomoniasis is the most common non-viral sexually transmitted infection caused by the vaginotropic extracellular protozoan parasite Trichomonas vaginalis. The infection is recurrent, with no lasting immunity, often asymptomatic, and linked to pregnancy complications and risk of viral infection. The molecular mechanisms of immune evasion by the parasite are poorly understood. We demonstrate that galectin-1 and -3 are expressed by the human cervical and vaginal epithelial cells and act as pathogen-recognition receptors for the ceramide phosphoinositol glycan core (CPI-GC) of the dominant surface protozoan lipophosphoglycan (LPG). We used an in vitro model with siRNA galectin knockdown epithelial clones, recombinant galectins, clinical Trichomonas isolates, and mutant protozoan derivatives to dissect the function of galectin-1 and -3 in the context of Trichomonas infection. Galectin-1 suppressed chemokines that facilitate recruitment of phagocytes, which can eliminate extracellular protozoa (IL-8) or bridge innate to adaptive immunity (MIP-3α and RANTES (regulated on activation normal T cell expressed and secreted)). Silencing galectin-1 increased and adding exogenous galectin-1 suppressed chemokine responses to Trichomonas or CPI-GC/LPG. In contrast, silencing galectin-3 reduced IL-8 response to LPG. Live Trichomonas depleted the extracellular levels of galectin-3. Clinical isolates and mutant Trichomonas CPI-GC that had reduced affinity to galectin-3 but maintained affinity to galectin-1 suppressed chemokine expression. Thus via CPI-GC binding, Trichomonas is capable of regulating galectin bioavailability and function to the benefit of its parasitic survival. These findings suggest novel approaches to control trichomoniasis and warrant further studies of galectin-binding diversity among clinical isolates as a possible source for symptom disparity in parasitic infections.
Collapse
Affiliation(s)
- Raina N Fichorova
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115,
| | - Hidemi S Yamamoto
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Titilayo Fashemi
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Evan Foley
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Stanthia Ryan
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Noah Beatty
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Hassan Dawood
- From the Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Gary R Hayes
- the Departments of Biochemistry and Molecular Biology and Obstetrics and Gynecology, State University of New York Upstate Medical University, Syracuse, New York 13210, and
| | - Guillaume St-Pierre
- the Laboratory of Glycobiology and Bioimaging, Research Centre for Infectious Diseases, Faculty of Medicine, Laval University, Quebec, Quebec G1V 4G2, Canada
| | - Sachiko Sato
- the Laboratory of Glycobiology and Bioimaging, Research Centre for Infectious Diseases, Faculty of Medicine, Laval University, Quebec, Quebec G1V 4G2, Canada
| | - Bibhuti N Singh
- the Departments of Biochemistry and Molecular Biology and Obstetrics and Gynecology, State University of New York Upstate Medical University, Syracuse, New York 13210, and
| |
Collapse
|
26
|
Vasta GR, Feng C, Bianchet MA, Bachvaroff TR, Tasumi S. Structural, functional, and evolutionary aspects of galectins in aquatic mollusks: From a sweet tooth to the Trojan horse. FISH & SHELLFISH IMMUNOLOGY 2015; 46:94-106. [PMID: 25982395 PMCID: PMC4509915 DOI: 10.1016/j.fsi.2015.05.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 04/30/2015] [Accepted: 05/04/2015] [Indexed: 05/02/2023]
Abstract
Galectins constitute a conserved and widely distributed lectin family characterized by their binding affinity for β-galactosides and a unique binding site sequence motif in the carbohydrate recognition domain (CRD). In spite of their structural conservation, galectins display a remarkable functional diversity, by participating in developmental processes, cell adhesion and motility, regulation of immune homeostasis, and recognition of glycans on the surface of viruses, bacteria and protozoan parasites. In contrast with mammals, and other vertebrate and invertebrate taxa, the identification and characterization of bona fide galectins in aquatic mollusks has been relatively recent. Most of the studies have focused on the identification and domain organization of galectin-like transcripts or proteins in diverse tissues and cell types, including hemocytes, and their expression upon environmental or infectious challenge. Lectins from the eastern oyster Crassostrea virginica, however, have been characterized in their molecular, structural and functional aspects and some notable features have become apparent in the galectin repertoire of aquatic mollusks. These including less diversified galectin repertoires and different domain organizations relative to those observed in vertebrates, carbohydrate specificity for blood group oligosaccharides, and up regulation of galectin expression by infectious challenge, a feature that supports their proposed role(s) in innate immune responses. Although galectins from some aquatic mollusks have been shown to recognize microbial pathogens and parasites and promote their phagocytosis, they can also selectively bind to phytoplankton components, suggesting that they also participate in uptake and intracellular digestion of microalgae. In addition, the experimental evidence suggests that the protozoan parasite Perkinsus marinus has co-evolved with the oyster host to be selectively recognized by the oyster hemocyte galectins over algal food or bacterial pathogens, thereby subverting the oyster's innate immune/feeding recognition mechanisms to gain entry into the host cells.
Collapse
Affiliation(s)
- G R Vasta
- Department of Microbiology and Immunology, University of Maryland School of Medicine, and Institute of Marine and Environmental Technology, Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA.
| | - C Feng
- Department of Microbiology and Immunology, University of Maryland School of Medicine, and Institute of Marine and Environmental Technology, Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA
| | - M A Bianchet
- Department of Neurology, and Department of Biophysics & Biophysical Chemistry, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - T R Bachvaroff
- University of Maryland Center for Environmental Science, and Institute of Marine and Environmental Technology, Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA
| | - S Tasumi
- Department of Microbiology and Immunology, University of Maryland School of Medicine, and Institute of Marine and Environmental Technology, Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA
| |
Collapse
|
27
|
Yuan C, Zhang H, Wang W, Li Y, Yan R, Xu L, Song X, Li X. Transmembrane protein 63A is a partner protein of Haemonchus contortus galectin in the regulation of goat peripheral blood mononuclear cells. Parasit Vectors 2015; 8:211. [PMID: 25879191 PMCID: PMC4404006 DOI: 10.1186/s13071-015-0816-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/17/2015] [Indexed: 01/04/2023] Open
Abstract
Background Hco-gal-m and -f were two isoforms of galectin cloned from male and female Haemonchus contortus, respectively, and it was demonstrated that recombinant Hco-gal-m and -f could act as immune suppressors. However, little is known about the receptors or binding partners of these galectins in the host. The research of the molecular mechanisms that govern the interactions between these galectins and host molecules will fill a gap in our understanding how parasite galectins interact with host cells. Methods A yeast two-hybrid system was used to identify the binding partners of Hco-gal-m and -f in this research. The interaction between rHco-gal-m and candidate binding protein was validated by co-immunoprecipitation. The localization of transmembrane protein 63A (TMEM63A) in peripheral blood mononuclear cells (PBMCs) was detected by immunofluorescence. The distribution of TMEM63A in T cells, B cells and monocytes in PBMCs was detected by flow cytometry. The immunomodulatory effects of Hco-gal-m and TMEM63A on cell proliferation, migration, apoptosis, nitric oxide production and cytokine secretion were observed by co-incubation of rHco-gal-m and TMEM63A-siRNA with goat PBMCs and monocytes. Results We found that TMEM63A, a functionally unknown protein, from goat PBMCs could bind to Hco-gal-m and -f. Immunofluorescence showed that TMEM63A was localized to the cell membrane. Flow cytometric analysis revealed that TMEM63A was expressed in the majority of goat PBMCs. After using RNA interference to knockdown expression of TMEM63A, the PBMC proliferation and migration were significantly increased, while the influence of rHco-gal-m on monocyte phagocytosis, PBMC nitric oxide production and migration were potently blocked. In addition, the production of IL-10, IFN-γ and TGF-β induced by rHco-gal-m were also altered. Conclusions Our results show that TMEM63A is a binding partner of Hco-gal-m/f, and involved in the immune responses of host PBMCs induced by Hco-gal-m for the first time. Electronic supplementary material The online version of this article (doi:10.1186/s13071-015-0816-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cheng Yuan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| | - Hui Zhang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| | - Wang Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| | - Yan Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| | - RuoFeng Yan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| | - LiXin Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| | - XiaoKai Song
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| | - XiangRui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| |
Collapse
|
28
|
Leishmania infantum amastigotes trigger a subpopulation of human B cells with an immunoregulatory phenotype. PLoS Negl Trop Dis 2015; 9:e0003543. [PMID: 25710789 PMCID: PMC4339978 DOI: 10.1371/journal.pntd.0003543] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 01/16/2015] [Indexed: 01/14/2023] Open
Abstract
Visceral leishmaniasis is caused by the protozoan parasites Leishmania infantum and Leishmania donovani. This infection is characterized by an uncontrolled parasitization of internal organs which, when left untreated, leads to death. Disease progression is linked with the type of immune response generated and a strong correlation was found between disease progression and serum levels of the immunosuppressive cytokine IL-10. Other studies have suggested a role for B cells in the pathology of this parasitic infection and the recent identification of a B-cell population in humans with regulatory functions, which secretes large amounts of IL-10 following activation, have sparked our interest in the context of visceral leishmaniasis. We report here that incubation of human B cells with Leishmania infantum amastigotes resulted in upregulation of multiple cell surface activation markers and a dose-dependent secretion of IL-10. Conditioned media from B cells incubated with Leishmania infantum amastigotes were shown to strongly inhibit CD4+ T-cell activation, proliferation and function (i.e. as monitored by TNF and IFNγ secretion). Blockade of IL-10 activity using a soluble IL-10 receptor restored only partially TNF and IFNγ production to control levels. The parasite-mediated IL-10 secretion was shown to rely on the activity of Syk, phosphatidylinositol-3 kinase and p38, as well as to require intracellular calcium mobilization. Cell sorting experiments allowed us to identify the IL-10-secreting B-cell subset (i.e. CD19+CD24+CD27-). In summary, exposure of human B cells to Leishmania infantum amastigotes triggers B cells with regulatory activities mediated in part by IL-10, which could favor parasite dissemination in the organism. Leishmaniasis is an infection caused by protozoan parasites of the genus Leishmania and is a significant neglected tropical disease, with 350 million people in 98 countries at risk of developing one of the forms of the disease. Visceral leishmaniasis is characterized by an uncontrolled parasitization of internal organs, which leads to death when left untreated. Disease progression is linked with the type of immune response generated and a strong correlation was found between disease progression and serum levels of the immunosuppressive cytokine IL-10. We demonstrate that a contact between human B cells with Leishmania infantum amastigotes resulted in upregulation of multiple cell surface activation markers and a dose-dependent secretion of IL-10. Conditioned media from B cells incubated with Leishmania infantum amastigotes were shown to strongly inhibit CD4+ T-cell activation, proliferation and function (i.e. TNF and IFNγ production). Blockade of IL-10 activity using a soluble IL-10 receptor restored to some degree TNF and IFNγ secretion. Cell sorting experiments allowed us to identify a major IL-10-secreting B cell subset characterized as CD24+ and CD27-. Exposure of human B cells to Leishmania infantum amastigotes thus triggers B cells with regulatory activities mediated in part by IL-10, which could promote parasite dissemination in the organism.
Collapse
|
29
|
Abstract
Recent reports suggest that some galectins bind to enveloped viruses. They include influenza virus, human immunodeficiency virus-1 (HIV-1), human T-cell leukemia virus-1 (HTLV-1), and Nipah virus. It is also suggested that the interaction between viruses and galectins influences viral attachment to their susceptible cells, affecting the viral infectivity. Our work suggests that galectin-1 increases the infectivity of HIV-1 and HTVL-1. Indeed, galectin-1 promotes the initial adsorption of HIV-1 to CD4(+) cells through its binding to viral envelope gp120 and facilitates HIV-1 infection in a manner that is dependent on its recognition of β-galactoside residues. Thus, as galectin-1 can be considered as a pattern recognition receptor, HIV-1 exploits this host factor to promote its transmission or replication. In this chapter, we describe methods used to investigate this potential role of galectins in HIV-1 infection as a case in point for future studies on galectin-virus interactions.
Collapse
Affiliation(s)
- Michel Ouellet
- Laboratory of Human Immuno-Retrovirology, Research Centre for Infectious Diseases, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | | | | | | |
Collapse
|
30
|
Preston S, Dunphy J, Beddoe T, Meeusen E, Young A. Evaluation of the role of galectins in parasite immunity. Methods Mol Biol 2015; 1207:371-95. [PMID: 25253154 DOI: 10.1007/978-1-4939-1396-1_25] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Galectin-11 and galectin-14 are ruminant galectins involved in parasitic infections. Although their roles in parasite immunity are still being elucidated, its appears that their functions are parasite specific. In gastrointestinal infections with the nematode Haemonchus contortus, both galectin-11 and galectin-14 appear to be protective. However, in a chronic infection of liver fluke, Fasciola hepatica, these galectins may aid parasite survival. This chapter discusses the methods designed to study parasitic infections in sheep, which have provided us with insight into the functions of galectin-11 and galectin-14 during host-parasite interactions. These methods include parasite cultivation and infection, galectin staining of host and parasite tissue, surface staining of parasites with recombinant galectins and in vitro assays to monitor the effect of galectins on larval development.
Collapse
Affiliation(s)
- Sarah Preston
- Biotechnology Research Laboratories, School of Biomedical Sciences, Monash University, Clayton, VIC, Australia
| | | | | | | | | |
Collapse
|
31
|
Abstract
Galectins are an evolutionarily ancient family of glycan-binding proteins (GBPs) and are found in all animals. Although they were discovered over 30 years ago, ideas about their biological functions continue to evolve. Current evidence indicates that galectins, which are the only known GBPs that occur free in the cytoplasm and extracellularly, are involved in a variety of intracellular and extracellular pathways contributing to homeostasis, cellular turnover, cell adhesion, and immunity. Here we review evolving insights into galectin biology from a historical perspective and explore current evidence regarding biological roles of galectins.
Collapse
|
32
|
Rauthu SR, Shiao TC, André S, Miller MC, Madej É, Mayo KH, Gabius HJ, Roy R. Defining the Potential of Aglycone Modifications for Affinity/Selectivity Enhancement against Medically Relevant Lectins: Synthesis, Activity Screening, and HSQC-Based NMR Analysis. Chembiochem 2014; 16:126-39. [DOI: 10.1002/cbic.201402474] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Indexed: 12/28/2022]
|
33
|
Pineda MA, Corvo L, Soto M, Fresno M, Bonay P. Interactions of human galectins with Trypanosoma cruzi: Binding profile correlate with genetic clustering of lineages. Glycobiology 2014; 25:197-210. [DOI: 10.1093/glycob/cwu103] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
34
|
Baum LG, Garner OB, Schaefer K, Lee B. Microbe-Host Interactions are Positively and Negatively Regulated by Galectin-Glycan Interactions. Front Immunol 2014; 5:284. [PMID: 24995007 PMCID: PMC4061488 DOI: 10.3389/fimmu.2014.00284] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 06/02/2014] [Indexed: 12/13/2022] Open
Abstract
Microbe–host interactions are complex processes that are directly and indirectly regulated by a variety of factors, including microbe presentation of specific molecular signatures on the microbial surface, as well as host cell presentation of receptors that recognize these pathogen signatures. Cell surface glycans are one important class of microbial signatures that are recognized by a variety of host cell lectins. Host cell lectins that recognize microbial glycans include members of the galectin family of lectins that recognize specific glycan ligands on viruses, bacteria, fungi, and parasites. In this review, we will discuss the ways that the interactions of microbial glycans with host cell galectins positively and negatively regulate pathogen attachment, invasion, and survival, as well as regulate host responses that mitigate microbial pathogenesis.
Collapse
Affiliation(s)
- Linda G Baum
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles , Los Angeles, CA , USA
| | - Omai B Garner
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles , Los Angeles, CA , USA
| | - Katrin Schaefer
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles , Los Angeles, CA , USA
| | - Benhur Lee
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles , Los Angeles, CA , USA ; Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at University of California Los Angeles , Los Angeles, CA , USA
| |
Collapse
|
35
|
Expression and function of galectins in the endometrium and at the human feto-maternal interface. Placenta 2013; 34:863-72. [PMID: 23911101 DOI: 10.1016/j.placenta.2013.07.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 07/08/2013] [Accepted: 07/11/2013] [Indexed: 12/12/2022]
Abstract
Galectins are classified as lectins that share structural similarities and bind β-galactosides via a conserved carbohydrate recognition domain. So far 16 out of 19 identified galectins were shown to be present in humans and numerous studies revealed galectins as pivotal modulators of cell death, differentiation and growth. Galectins were highlighted to interact with both the adaptive and innate immune response. In the field of reproductive medicine and placenta research different roles for galectins have been proposed. Several galectins, being abundantly present at the human feto-maternal interphase and endometrium, were hypothesized to significantly contribute to endometrial receptivity and pregnancy physiology. Hence, this review outlines selected aspects of galectin action within endometrial function and at the feto-maternal interphase. Further current knowledge on galectins in reproductive and pregnancy disorders like endometriosis, abortion or preeclampsia is summarized.
Collapse
|
36
|
Sohrabi Y, Havelková H, Kobets T, Šíma M, Volkova V, Grekov I, Jarošíková T, Kurey I, Vojtíšková J, Svobodová M, Demant P, Lipoldová M. Mapping the genes for susceptibility and response to Leishmania tropica in mouse. PLoS Negl Trop Dis 2013; 7:e2282. [PMID: 23875032 PMCID: PMC3708836 DOI: 10.1371/journal.pntd.0002282] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 05/09/2013] [Indexed: 12/04/2022] Open
Abstract
Background L. tropica can cause both cutaneous and visceral leishmaniasis in humans. Although the L. tropica-induced cutaneous disease has been long known, its potential to visceralize in humans was recognized only recently. As nothing is known about the genetics of host responses to this infection and their clinical impact, we developed an informative animal model. We described previously that the recombinant congenic strain CcS-16 carrying 12.5% genes from the resistant parental strain STS/A and 87.5% genes from the susceptible strain BALB/c is more susceptible to L. tropica than BALB/c. We used these strains to map and functionally characterize the gene-loci regulating the immune responses and pathology. Methods We analyzed genetics of response to L. tropica in infected F2 hybrids between BALB/c×CcS-16. CcS-16 strain carries STS-derived segments on nine chromosomes. We genotyped these segments in the F2 hybrid mice and tested their linkage with pathological changes and systemic immune responses. Principal Findings We mapped 8 Ltr (Leishmania tropica response) loci. Four loci (Ltr2, Ltr3, Ltr6 and Ltr8) exhibit independent responses to L. tropica, while Ltr1, Ltr4, Ltr5 and Ltr7 were detected only in gene-gene interactions with other Ltr loci. Ltr3 exhibits the recently discovered phenomenon of transgenerational parental effect on parasite numbers in spleen. The most precise mapping (4.07 Mb) was achieved for Ltr1 (chr.2), which controls parasite numbers in lymph nodes. Five Ltr loci co-localize with loci controlling susceptibility to L. major, three are likely L. tropica specific. Individual Ltr loci affect different subsets of responses, exhibit organ specific effects and a separate control of parasite load and organ pathology. Conclusion We present the first identification of genetic loci controlling susceptibility to L. tropica. The different combinations of alleles controlling various symptoms of the disease likely co-determine different manifestations of disease induced by the same pathogen in individual mice. Leishmaniasis, a disease caused by Leishmania ssp. is among the most neglected infectious diseases. In humans, L. tropica causes cutaneous form of leishmaniasis, but can damage internal organs too. The reasons for this variability are not known, and its genetic basis was never investigated. Therefore, analysis of genes affecting host's responses to this infection can elucidate the characteristics of individual host-parasite interactions. Recombinant congenic strain CcS-16 carries 12.5% genes from the mouse strain STS/A on genetic background of the strain BALB/c, and it is more susceptible than BALB/c. In F2 hybrids between BALB/c and CcS-16 we detected and mapped eight gene-loci, Ltr1-8 (Leishmania tropica response 1-8) that control various manifestations of disease: skin lesions, splenomegaly, hepatomegaly, parasite numbers in spleen, liver, and inguinal lymph nodes, and serum level of CCL3, CCL5, and CCL7 after L. tropica infection. These loci are functionally heterogeneous - each influences a different set of responses to the pathogen. Five loci co-localize with the previously described loci that control susceptibility to L. major, three are species-specific. Ltr2 co-localizes not only with Lmr14 (Leishmania major response 14), but also with Ir2 influencing susceptibility to L. donovani and might therefore carry a common gene controlling susceptibility to leishmaniasis.
Collapse
Affiliation(s)
- Yahya Sohrabi
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Helena Havelková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Tetyana Kobets
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Matyáš Šíma
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Valeriya Volkova
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Igor Grekov
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Taťána Jarošíková
- Faculty of Biomedical Engineering, Czech Technical University in Prague, Kladno, Czech Republic
| | - Iryna Kurey
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jarmila Vojtíšková
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | - Peter Demant
- Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Marie Lipoldová
- Laboratory of Molecular and Cellular Immunology, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
37
|
Heusschen R, Griffioen AW, Thijssen VL. Galectin-9 in tumor biology: a jack of multiple trades. Biochim Biophys Acta Rev Cancer 2013; 1836:177-85. [PMID: 23648450 DOI: 10.1016/j.bbcan.2013.04.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 04/25/2013] [Accepted: 04/29/2013] [Indexed: 12/22/2022]
Abstract
Galectin family members have been shown to exert multiple roles in the context of tumor biology. Several recent findings support a similar multi-faceted role for galectin-9. Galectin-9 expression is frequently altered in cancer as compared to normal tissues. In addition, an increasing amount of evidence suggests that galectin-9 is involved in several aspects of tumor progression, including tumor cell adhesion and survival, immune escape and angiogenesis. Also, galectin-9 shows potential as a prognostic marker and a therapeutic target for several malignancies. In this review we summarize both the established and the emerging roles of galectin-9 in tumor biology and discuss the potential application of galectin-9 in anti-cancer therapy.
Collapse
Affiliation(s)
- Roy Heusschen
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
38
|
Bhaumik P, St-Pierre G, Milot V, St-Pierre C, Sato S. Galectin-3 Facilitates Neutrophil Recruitment as an Innate Immune Response to a Parasitic Protozoa Cutaneous Infection. THE JOURNAL OF IMMUNOLOGY 2012; 190:630-40. [DOI: 10.4049/jimmunol.1103197] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Vasta GR, Ahmed H, Bianchet MA, Fernández-Robledo JA, Amzel LM. Diversity in recognition of glycans by F-type lectins and galectins: molecular, structural, and biophysical aspects. Ann N Y Acad Sci 2012; 1253:E14-26. [PMID: 22973821 DOI: 10.1111/j.1749-6632.2012.06698.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Although lectins are "hard-wired" in the germline, the presence of tandemly arrayed carbohydrate recognition domains (CRDs), of chimeric structures displaying distinct CRDs, of polymorphic genes resulting in multiple isoforms, and in some cases, of a considerable recognition plasticity of their carbohydrate binding sites, significantly expand the lectin ligand-recognition spectrum and lectin functional diversification. Analysis of structural/functional aspects of galectins and F-lectins-the most recently identified lectin family characterized by a unique CRD sequence motif (a distinctive structural fold) and nominal specificity for l-Fuc-has led to a greater understanding of self/nonself recognition by proteins with tandemly arrayed CRDs. For lectins with a single CRD, however, recognition of self and nonself glycans can only be rationalized in terms of protein oligomerization and ligand clustering and presentation. Spatial and temporal changes in lectin expression, secretion, and local concentrations in extracellular microenvironments, as well as structural diversity and spatial display of their carbohydrate ligands on the host or microbial cell surface, are suggestive of a dynamic interplay of their recognition and effector functions in development and immunity.
Collapse
Affiliation(s)
- Gerardo R Vasta
- Department of Microbiology and Immunology, University of Maryland School of Medicine, IMET, Baltimore, Maryland 21202-3101, USA.
| | | | | | | | | |
Collapse
|
40
|
Liu D, Uzonna JE. The early interaction of Leishmania with macrophages and dendritic cells and its influence on the host immune response. Front Cell Infect Microbiol 2012; 2:83. [PMID: 22919674 PMCID: PMC3417671 DOI: 10.3389/fcimb.2012.00083] [Citation(s) in RCA: 220] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 05/27/2012] [Indexed: 01/26/2023] Open
Abstract
The complicated interactions between Leishmania and the host antigen-presenting cells (APCs) have fundamental effects on the final outcome of the disease. Two major APCs, macrophages and dendritic cells (DCs), play critical roles in mediating resistance and susceptibility during Leishmania infection. Macrophages are the primary resident cell for Leishmania: they phagocytose and permit parasite proliferation. However, these cells are also the major effector cells to eliminate infection. The effective clearance of parasites by macrophages depends on activation of appropriate immune response, which is usually initiated by DCs. Here, we review the early interaction of APCs with Leishmania parasites and how these interactions profoundly impact on the ensuing adaptive immune response. We also discuss how the current knowledge will allow further refinement of our understanding of the interplay between Leishmania and its hosts that leads to resistance or susceptibility.
Collapse
Affiliation(s)
- Dong Liu
- Department of Immunology, University of Manitoba, Winnipeg MB, Canada
| | | |
Collapse
|
41
|
Elahi S, Niki T, Hirashima M, Horton H. Galectin-9 binding to Tim-3 renders activated human CD4+ T cells less susceptible to HIV-1 infection. Blood 2012; 119:4192-204. [PMID: 22438246 PMCID: PMC3359739 DOI: 10.1182/blood-2011-11-389585] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 02/16/2012] [Indexed: 01/19/2023] Open
Abstract
Galectin-9 (Gal-9) is a tandem repeat-type member of the galectin family and is a ligand for T-cell immunoglobulin mucin domain 3 (Tim-3), a type-I glycoprotein that is persistently expressed on dysfunctional T cells during chronic infection. Studies in autoimmune diseases and chronic viral infections show that Tim-3 is a regulatory molecule that inhibits Th1 type immune responses. Here we show that soluble Gal-9 interacts with Tim-3 expressed on the surface of activated CD4(+) T cells and renders them less susceptible to HIV-1 infection and replication. The Gal-9/Tim-3 interaction on activated CD4(+) T cells, leads to down-regulation of HIV-1 coreceptors and up-regulation of the cyclin-dependent kinase inhibitor p21 (also known as cip-1 and waf-1). We suggest that higher expression of Tim-3 during chronic infection has evolved to limit persistent immune activation and associated tissue damage. These data demonstrate a novel mechanism for Gal-9/Tim-3 interactions to induce resistance of activated CD4(+) T cells to HIV-1 infection and suggest that Gal-9 may play a role in HIV-1 pathogenesis and could be used as a novel microbicide to prevent HIV-1 infection.
Collapse
Affiliation(s)
- Shokrollah Elahi
- Viral Vaccine Program, Seattle Biomedical Research Institute (Seattle Biomed), 307 Westlake Ave N, Seattle, WA 98109, USA
| | | | | | | |
Collapse
|
42
|
Sato S, Ouellet M, St-Pierre C, Tremblay MJ. Glycans, galectins, and HIV-1 infection. Ann N Y Acad Sci 2012; 1253:133-48. [DOI: 10.1111/j.1749-6632.2012.06475.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
43
|
Galectin-1-specific inhibitors as a new class of compounds to treat HIV-1 infection. Antimicrob Agents Chemother 2011; 56:154-62. [PMID: 22064534 DOI: 10.1128/aac.05595-11] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Despite significant improvements, antiretroviral therapies against HIV-1 are plagued by a high frequency of therapeutic failures that have been associated with acquisition of drug resistance. We recently reported that HIV-1 exploits a host glycan binding protein, galectin-1, to increase its attachment to host cells, thereby increasing its overall infectivity in susceptible cells. This finding suggests that host molecules such as galectin-1 could reduce the expected efficiency of HIV-1 drugs targeting early steps of the replicative cycle, such as attachment and entry processes. Thus, new classes of drugs that would interfere with galectin-1/HIV-1 interactions could benefit the current antiretroviral therapy. To further explore this possibility, experiments were conducted to discover leading compounds showing specific inhibition of galectin-1 activity in a cellular model of HIV-1 infection. Three lactoside compounds were found to modestly inhibit the interaction of galectin-1 with primary human CD4(+) T cells. Interestingly, these same inhibitors reduced the galectin-1-mediated increase in HIV-1 attachment to target cells in a much more efficient manner. More important, the tested lactoside derivatives also significantly decreased the galectin-1-dependent enhancement of HIV-1 infection. These observations deserve further attention when considering that the development of new drugs to prevent and treat HIV-1 infection remains a priority.
Collapse
|
44
|
Host-soluble galectin-1 promotes HIV-1 replication through a direct interaction with glycans of viral gp120 and host CD4. J Virol 2011; 85:11742-51. [PMID: 21880749 DOI: 10.1128/jvi.05351-11] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sexual transmission of HIV-1 requires virus adsorption to a target cell, typically a CD4(+) T lymphocyte residing in the lamina propria, beneath the epithelium. To escape the mucosal clearance system and reach its target cells, HIV-1 has evolved strategies to circumvent deleterious host factors. Galectin-1, a soluble lectin found in the underlayers of the epithelium, increases HIV-1 infectivity by accelerating its binding to susceptible cells. By comparison, galectin-3, a family member expressed by epithelial cells and part of the mucosal clearance system, does not perform similarly. We show here that galectin-1 directly binds to HIV-1 in a β-galactoside-dependent fashion through recognition of clusters of N-linked glycans on the viral envelope gp120. Unexpectedly, this preferential binding of galectin-1 does not rely on the primary sequence of any particular glycans. Instead, glycan clustering arising from the tertiary structure of gp120 hinders its binding by galectin-3. Increased polyvalency of a specific ligand epitope is a common strategy for glycans to increase their avidity for lectins. In this peculiar occurrence, glycan clustering is instead exploited to prevent binding of gp120 by galectin-3, which would lead to a biological dead-end for the virus. Our data also suggest that galectin-1 binds preferentially to CD4, the host receptor for gp120. Together, these results suggest that HIV-1 exploits galectin-1 to enhance gp120-CD4 interactions, thereby promoting virus attachment and infection events. Since viral adhesion is a rate-limiting step for HIV-1 entry, modulation of the gp120 interaction with galectin-1 could thus represent a novel approach for the prevention of HIV-1 transmission.
Collapse
|
45
|
Wiersma VR, de Bruyn M, Helfrich W, Bremer E. Therapeutic potential of Galectin-9 in human disease. Med Res Rev 2011; 33 Suppl 1:E102-26. [PMID: 21793015 DOI: 10.1002/med.20249] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In recent years, an important role has emerged for the glycan-binding protein Galectin-9 (Gal-9) in health and disease. In normal physiology, Gal-9 seems to be a pivotal modulator of T-cell immunity by inducing apoptosis in specific T-cell subpopulations. Because these T-cell populations are associated with autoimmunity, inflammatory disease, and graft rejection, it was postulated that application of exogenous Gal-9 may limit pathogenic T-cell activity. Indeed, treatment with recombinant Gal-9 ameliorates disease activity in various preclinical models of autoimmunity and allograft graft rejection. In many solid cancers, the loss of Gal-9 expression is closely associated with metastatic progression. In line with this observation, treatment with recombinant Gal-9 prevents metastatic spread in various preclinical cancer models. In addition, various hematological malignancies are sensitive to apoptotic elimination by recombinant Gal-9. Here, we review the biology and physiological role of this versatile lectin and discuss the therapeutic potential of Gal-9 in various diseases, including autoimmunity, asthma, infection, and cancer.
Collapse
Affiliation(s)
- Valerie R Wiersma
- Department of Surgery, Surgical Research Laboratories, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | |
Collapse
|
46
|
Song X, Zhang H, Wang L, Zhao J, Mu C, Song L, Qiu L, Liu X. A galectin with quadruple-domain from bay scallop Argopecten irradians is involved in innate immune response. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:592-602. [PMID: 21237194 DOI: 10.1016/j.dci.2011.01.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 01/07/2011] [Accepted: 01/07/2011] [Indexed: 05/30/2023]
Abstract
Galectins are a family of β-galactoside-binding lectins that specifically bind to β-galactoside residues and play crucial roles in innate immune responses of invertebrates and vertebrates. The cDNA of bay scallop Argopecten irradians galectin (designated as AiGal2) was cloned by rapid amplification of cDNA ends (RACE) method based on the expressed sequence tag (EST). The full-length cDNA of AiGal2 was of 2137 bp. The open reading frame encoded a polypeptide of 555 amino acids containing four carbohydrate-recognition domains. The deduced amino acid sequence and multi-domain organization of AiGal2 were highly similar to those of mollusk galectins. A typical galectin fold in β-sandwich arrangement was identified in the potential tertiary structure of all the four CRDs in AiGal2. The mRNA transcripts of AiGal2 were found to be constitutively expressed in a wide range of tissues and mainly in hepatopancreas, adductor muscle and kidney. After scallops were challenged by Vibrio anguillarum or Micrococcus luteus, the mRNA expression level of AiGal2 was up-regulated significantly, while it did not changed remarkably after Pichia pastoris challenge. The recombined AiGal2 (rAiGal2) exhibited strong activity to agglutinate E. coli, V. anguillarum, Vibrio fluvialis, Edwardsiella tarda and M. luteus, and the agglutinating activities could be inhibited by both d-galactose and lactose. The in vitro encapsulation assay revealed that rAiGal2 could bind to hemocytes and enhanced its encapsulation of agarose beads. These results collectively suggested that AiGal2 functioned as a pattern recognition receptor in immune defense and contributed to the non-self recognition and elimination in cellular immune response of bay scallop.
Collapse
Affiliation(s)
- Xiaoyan Song
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Cerliani JP, Stowell SR, Mascanfroni ID, Arthur CM, Cummings RD, Rabinovich GA. Expanding the universe of cytokines and pattern recognition receptors: galectins and glycans in innate immunity. J Clin Immunol 2010; 31:10-21. [PMID: 21184154 DOI: 10.1007/s10875-010-9494-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 12/03/2010] [Indexed: 12/31/2022]
Abstract
Effective immunity relies on the recognition of pathogens and tumors by innate immune cells through diverse pattern recognition receptors (PRRs) that lead to initiation of signaling processes and secretion of pro- and anti-inflammatory cytokines. Galectins, a family of endogenous lectins widely expressed in infected and neoplastic tissues have emerged as part of the portfolio of soluble mediators and pattern recognition receptors responsible for eliciting and controlling innate immunity. These highly conserved glycan-binding proteins can control immune cell processes through binding to specific glycan structures on pathogens and tumors or by acting intracellularly via modulation of selective signaling pathways. Recent findings demonstrate that various galectin family members influence the fate and physiology of different innate immune cells including polymorphonuclear neutrophils, mast cells, macrophages, and dendritic cells. Moreover, several pathogens may actually utilize galectins as a mechanism of host invasion. In this review, we aim to highlight and integrate recent discoveries that have led to our current understanding of the role of galectins in host-pathogen interactions and innate immunity. Challenges for the future will embrace the rational manipulation of galectin-glycan interactions to instruct and shape innate immunity during microbial infections, inflammation, and cancer.
Collapse
Affiliation(s)
- Juan P Cerliani
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, 1428 Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
48
|
Debierre-Grockiego F, Niehus S, Coddeville B, Elass E, Poirier F, Weingart R, Schmidt RR, Mazurier J, Guérardel Y, Schwarz RT. Binding of Toxoplasma gondii glycosylphosphatidylinositols to galectin-3 is required for their recognition by macrophages. J Biol Chem 2010; 285:32744-32750. [PMID: 20729207 DOI: 10.1074/jbc.m110.137588] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We showed that the production of tumor necrosis factor (TNF) α by macrophages in response to Toxoplasma gondii glycosylphosphatidylinositols (GPIs) requires the expression of both Toll-like receptors TLR2 and TLR4, but not of their co-receptor CD14. Galectin-3 is a β-galactoside-binding protein with immune-regulatory effects, which associates with TLR2. We demonstrate here by using the surface plasmon resonance method that the GPIs of T. gondii bind to human galectin-3 with strong affinity and in a dose-dependent manner. The use of a synthetic glycan and of the lipid moiety cleaved from the GPIs shows that both parts are involved in the interaction with galectin-3. GPIs of T. gondii also bind to galectin-1 but with a lower affinity and only through the lipid moiety. At the cellular level, the production of TNF-α induced by T. gondii GPIs in macrophages depends on the expression of galectin-3 but not of galectin-1. This study is the first identification of a galectin-3 ligand of T. gondii origin, and galectin-3 might be a co-receptor presenting the GPIs to the TLRs on macrophages.
Collapse
Affiliation(s)
- Françoise Debierre-Grockiego
- From the Institut für Virologie, AG Parasitologie, Philipps University, Marburg D-35043, Germany; Unité Mixte de Recherche Université-Institut National de la Recherche Agronomique 0483 Immunologie Parasitaire, Vaccinologie et Biothérapies Anti-infectieuses, Unité de Formation de Recherche Sciences Pharmaceutiques, 31 Avenue Monge, F-37200 Tours, France.
| | - Sebastian Niehus
- From the Institut für Virologie, AG Parasitologie, Philipps University, Marburg D-35043, Germany
| | - Bernadette Coddeville
- CNRS-Unité Mixte de Recherche 8576, Unit of Structural and Functional Glycobiology, Institut Fédératif de Recherche 147, Université Lille Nord de France, F-59000 Lille, France
| | - Elisabeth Elass
- CNRS-Unité Mixte de Recherche 8576, Unit of Structural and Functional Glycobiology, Institut Fédératif de Recherche 147, Université Lille Nord de France, F-59000 Lille, France
| | - Françoise Poirier
- Laboratoire de Génétique et Développement des Mammifères, Institut Jacques Monod, Paris F-75013, France
| | - Ralf Weingart
- Fachbereich Chemie, University of Konstanz, Konstanz D-78457, Germany
| | - Richard R Schmidt
- Fachbereich Chemie, University of Konstanz, Konstanz D-78457, Germany
| | - Joël Mazurier
- CNRS-Unité Mixte de Recherche 8576, Unit of Structural and Functional Glycobiology, Institut Fédératif de Recherche 147, Université Lille Nord de France, F-59000 Lille, France
| | - Yann Guérardel
- CNRS-Unité Mixte de Recherche 8576, Unit of Structural and Functional Glycobiology, Institut Fédératif de Recherche 147, Université Lille Nord de France, F-59000 Lille, France
| | - Ralph T Schwarz
- From the Institut für Virologie, AG Parasitologie, Philipps University, Marburg D-35043, Germany; CNRS-Unité Mixte de Recherche 8576, Unit of Structural and Functional Glycobiology, Institut Fédératif de Recherche 147, Université Lille Nord de France, F-59000 Lille, France
| |
Collapse
|
49
|
Song X, Zhang H, Zhao J, Wang L, Qiu L, Mu C, Liu X, Qiu L, Song L. An immune responsive multidomain galectin from bay scallop Argopectens irradians. FISH & SHELLFISH IMMUNOLOGY 2010; 28:326-332. [PMID: 19944171 DOI: 10.1016/j.fsi.2009.11.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 10/21/2009] [Accepted: 11/14/2009] [Indexed: 05/28/2023]
Abstract
Galectins are a family of beta-galactoside-binding lectins which play crucial roles in innate immunity of vertebrates and invertebrates. In the present study, the cDNA of a galectin with multiple carbohydrate-recognition domains (CRDs) was cloned from bay scallop Argopectens irradians (designated AiGal1) by expressed sequence tag (EST) and rapid amplification of cDNA ends (RACE) techniques. The full-length cDNA of AiGal1 was of 2235 nucleotides, encoding a polypeptide of 549 amino acids. SMART program analysis revealed that AiGal1 contained four galectin CRDs, and all the CRDs contained the two consensus motifs essential for ligand-binding. Quantitative real-time PCR was employed to investigate the tissue distribution of AiGal1 mRNA and temporal expression in haemocytes of scallops challenged with Vibrio anguillarum, Micrococcus luteus and Pichia pastoris. The AiGal1 mRNA could be detected in all tested tissues with the highest expression level in hepatopancreas. After challenged by V. anguillarum and M. luteus, the expression level of AiGal1 mRNA was both up-regulated and reached the maximum level at 9 h (1.52 fold, P < 0.05) and 18 h (2.89 fold, P < 0.01) post challenge, respectively. However, there was no significant difference in the mRNA expression of AiGal1 in haemocytes after P. pastoris challenge (P > 0.05). These results collectively indicated that AiGal1 was a new member of the galectin family and involved in the immune responses against bacterial infection.
Collapse
Affiliation(s)
- Xiaoyan Song
- The Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Initial binding of human immunodeficiency virus-1 (HIV-1) to its susceptible CD4(+) cells is the limiting step for the establishment of infection as the avidity of viral envelope gp120 for CD4 is not high and the number of viral envelope spikes on the surface is found to be low compared to highly infectious viruses. Several host factors, such as C-type lectins, are listed as being able to enforce or facilitate the crucial interaction of HIV-1 to the susceptible cell. Recent works suggest that a host soluble beta-galactoside-binding lectin, galectin-1, also facilitates both virion binding and the infection of target cells in a manner dependent on lactose but not mannose, suggesting that this soluble galectin can be considered as a host factor that influences HIV-1 pathogenesis. In this chapter, we describe methods used to investigate the potential role of the galectin family in HIV-1-mediated disease progression.
Collapse
Affiliation(s)
- Christian St-Pierre
- Glycobiology and Bioimaging Laboratory, Research Centre for Infectious Diseases, Faculty of Medicine, Laval University, Quebec, Canada
| | | | | | | |
Collapse
|