1
|
Malamud M, Brown GD. The Dectin-1 and Dectin-2 clusters: C-type lectin receptors with fundamental roles in immunity. EMBO Rep 2024:10.1038/s44319-024-00296-2. [PMID: 39482490 DOI: 10.1038/s44319-024-00296-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024] Open
Abstract
The ability of myeloid cells to recognize and differentiate endogenous or exogenous ligands rely on the presence of different transmembrane protein receptors. C-type lectin receptors (CLRs), defined by the presence of a conserved structural motif called C-type lectin-like domain (CTLD), are a crucial family of receptors involved in this process, being able to recognize a diverse range of ligands from glycans to proteins or lipids and capable of initiating an immune response. The Dectin-1 and Dectin-2 clusters involve two groups of CLRs, with genes genomically linked within the natural killer cluster of genes in both humans and mice, and all characterized by the presence of a single extracellular CTLD. Fundamental immune cell functions such as antimicrobial effector mechanisms as well as internalization and presentation of antigens are induced and/or regulated through activatory, or inhibitory signalling pathways triggered by these receptors after ligand binding. In this review, we will discuss the most recent concepts regarding expression, ligands, signaling pathways and functions of each member of the Dectin clusters of CLRs, highlighting the importance and diversity of their functions.
Collapse
Affiliation(s)
- Mariano Malamud
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| | - Gordon D Brown
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| |
Collapse
|
2
|
Metwali N, Stapleton EM, Hadina S, Thorne PS. Exposure to structurally unique β-d-glucans differentially affects inflammatory responses in male mouse lungs. Physiol Rep 2024; 12:e16115. [PMID: 38923221 PMCID: PMC11194181 DOI: 10.14814/phy2.16115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Pro-inflammatory fungal β-d-glucan (BDG) polysaccharides cause respiratory pathology. However, specific immunological effects of unique BDG structures on pulmonary inflammation are understudied. We characterized the effect of four unique fungal BDGs with unique branching patterns, solubility, and molecular weights in murine airways. Scleroglucan (1 → 3)(1 → 6)-highly branched BDG, laminarin (1 → 3)(1 → 6)-branched BDG, curdlan (1 → 3)-linear BDG, and pustulan (1 → 6)-linear BDG were assessed by nuclear magnetic resonance spectroscopy. Each BDG was tested by inhalation model with C3HeB/FeJ mice and compared to saline-exposed control mice and unexposed sentinels (n = 3-19). Studies were performed ±heat-inactivation (1 h autoclave) to increase BDG solubility. Outcomes included bronchoalveolar lavage (BAL) differential cell counts (macrophages, neutrophils, lymphocytes, eosinophils), cytokines, serum IgE, and IgG2a (multiplex and ELISA). Ex vivo primary cells removed from lungs and plated at monolayer were stimulated (BDG, lipopolysaccharide (LPS), anti-CD3), and cytokines compared to unstimulated cells. Right lung histology was performed. Inhalation of BDGs with distinct branching patterns exhibited varying inflammatory potency and immunogenicity. Lichen-derived (1 → 6)-linear pustulan was the most pro-inflammatory BDG, increasing inflammatory infiltrate (BAL), serum IgE and IgG2a, and cytokine production. Primed lung cells responded to secondary LPS stimulation with a T-cell-specific response to pustulan. Glucan source and solubility should be considered in exposure and toxicological studies.
Collapse
Affiliation(s)
- Nervana Metwali
- Department of Occupational and Environmental HealthCollege of Public Health, University of IowaIowa CityIowaUSA
| | - Emma M. Stapleton
- Division of Pulmonary Critical Care and Occupational Medicine, Department of Internal MedicineUniversity of IowaIowa CityIowaUSA
| | - Suzana Hadina
- Department of Occupational and Environmental HealthCollege of Public Health, University of IowaIowa CityIowaUSA
- Present address:
Department of Microbiology & Infectious Disease with ClinicFaculty of Veterinary Medicine, University of ZagrebZagrebCroatia
| | - Peter S. Thorne
- Department of Occupational and Environmental HealthCollege of Public Health, University of IowaIowa CityIowaUSA
| |
Collapse
|
3
|
Martínez-López S, Ángel-Gomis E, Gómez-Hurtado I, Fernández-Iglesias A, Morante J, Gracia-Sancho J, Boix P, Cubero FJ, Zapater P, Caparrós E, Francés R. Cirrhosis-downregulated LSECtin can be retrieved by cytokines, shifts the TLR-induced LSECs secretome and correlates with the hepatic Th response. Liver Int 2024; 44:996-1010. [PMID: 38293766 DOI: 10.1111/liv.15836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/07/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND AND AIMS We evaluated tolerogenic C-type lectin LSECtin loss in cirrhosis and its potential regulation by cytokines. METHODS Liver tissue from patients with cirrhosis and healthy controls, immortalised and generated LSECtin-CRISPR immortalised LSECs, and murine primary LSECs from the CCl4 model were handled. RESULTS LSECtin expression was reduced in liver tissue from cirrhotic patients, and it decreased from compensated to decompensated disease. Increased phosphorylation of MAPK, Akt and NFkB was observed upon LSECtin stimulation in LSEC murine cell line, showing a pattern of inflammatory and chemotactic cytokines either restrained (IL-10, CCL4) or unrestrained (TNF-α, IL-1β, IL-6, CCL2). CD44 attenuated whereas LAG-3 increased all substrates phosphorylation in combination with TLR4 and TLR2 ligands except for NFkB. TNF-α, IL-1 β, IL-6 and CCL2 were restrained by LSECtin crosslinking on TLRs studied. Conversely, IL-10 and CCL4 were upregulated, suggesting a LSECtin-TLRs synergistic effect. Also, LSECtin was significantly induced after IL-13 stimulation or combined with anti-inflammatory cytokines in cirrhotic and immortalised LSECs. Th17 and regulatory T cells were progressively increased in the hepatic tissue from compensated to decompensated patients. A significant inverse correlation was present between gene expression levels of CLEC4G/LSECtin and RORγT and FOXP3 in liver tissues. CONCLUSION LSECtin restrains TLR proinflammatory secretome induced on LSECs by interfering immune response control, survival and MAPKs signalling pathways. The cytokine-dependent induction of LSECtin and the association between LSECtin loss and Th17 cell subset expansion in the liver, provides a solid background for exploring LSECtin retrieval as a mechanism to reprogram LSEC homeostatic function hampered during cirrhosis.
Collapse
Affiliation(s)
- Sebastián Martínez-López
- Hepatic and Intestinal Immunobiology Group, Departamento de Medicina Clínica, Universidad Miguel Hernández, San Juan de Alicante, Spain
- IIS ISABIAL, Hospital General Universitario Dr. Balmis, Alicante, Spain
| | - Enrique Ángel-Gomis
- Hepatic and Intestinal Immunobiology Group, Departamento de Medicina Clínica, Universidad Miguel Hernández, San Juan de Alicante, Spain
- IIS ISABIAL, Hospital General Universitario Dr. Balmis, Alicante, Spain
| | - Isabel Gómez-Hurtado
- Hepatic and Intestinal Immunobiology Group, Departamento de Medicina Clínica, Universidad Miguel Hernández, San Juan de Alicante, Spain
- IIS ISABIAL, Hospital General Universitario Dr. Balmis, Alicante, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | - Anabel Fernández-Iglesias
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Liver Vascular Biology Research Group, IDIBAPS, Barcelona, Spain
| | - Javier Morante
- Instituto de Neurociencias, CSIC-UMH, San Juan de Alicante, Spain
| | - Jordi Gracia-Sancho
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Liver Vascular Biology Research Group, IDIBAPS, Barcelona, Spain
| | - Paula Boix
- Hepatic and Intestinal Immunobiology Group, Departamento de Medicina Clínica, Universidad Miguel Hernández, San Juan de Alicante, Spain
- IIS ISABIAL, Hospital General Universitario Dr. Balmis, Alicante, Spain
| | - Francisco J Cubero
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University, Madrid, Spain
| | - Pedro Zapater
- Hepatic and Intestinal Immunobiology Group, Departamento de Medicina Clínica, Universidad Miguel Hernández, San Juan de Alicante, Spain
- IIS ISABIAL, Hospital General Universitario Dr. Balmis, Alicante, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Instituto IDIBE, Universidad Miguel Hernández, Elche, Spain
| | - Esther Caparrós
- Hepatic and Intestinal Immunobiology Group, Departamento de Medicina Clínica, Universidad Miguel Hernández, San Juan de Alicante, Spain
- IIS ISABIAL, Hospital General Universitario Dr. Balmis, Alicante, Spain
| | - Rubén Francés
- Hepatic and Intestinal Immunobiology Group, Departamento de Medicina Clínica, Universidad Miguel Hernández, San Juan de Alicante, Spain
- IIS ISABIAL, Hospital General Universitario Dr. Balmis, Alicante, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
- Instituto IDIBE, Universidad Miguel Hernández, Elche, Spain
| |
Collapse
|
4
|
Torigoe S, Lowman DW, Sugiki T, Williams DL, Yamasaki S. Self-recognition through Dectin-1 exacerbates liver inflammation. Genes Cells 2024; 29:316-327. [PMID: 38385597 PMCID: PMC11000461 DOI: 10.1111/gtc.13106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/03/2024] [Accepted: 02/04/2024] [Indexed: 02/23/2024]
Abstract
Dectin-1 is a well-characterized C-type lectin receptor involved in anti-fungal immunity through the recognition of polysaccharides; however, molecular mechanisms and outcomes initiated through self-recognition have not been fully understood. Here, we purified a water-soluble fraction from mouse liver that acts as a Dectin-1 agonist. To address the physiological relevance of this recognition, we utilized sterile liver inflammation models. The CCl4-induced hepatitis model showed that Dectin-1 deficiency led to reduced inflammation through decreased inflammatory cell infiltration and lower pro-inflammatory cytokine levels. Moreover, in a NASH model induced by streptozotocin and a high-fat diet, hepatic inflammation and fibrosis were ameliorated in Dectin-1-deficient mice. The Dectin-1 agonist activity was increased in the water-soluble fraction from NASH mice, suggesting a potential pathogenic cycle between Dectin-1 activation and hepatitis progression. In vivo administration of the fraction into mice induced hepatic inflammation. These results highlight a role of self-recognition through Dectin-1 that triggers hepatic innate immune responses and contributes to the exacerbation of inflammation in pathogenic settings. Thus, the blockade of this axis may provide a therapeutic option for liver inflammatory diseases.
Collapse
Affiliation(s)
- Shota Torigoe
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo, Japan
- Management Department of Biosafety, Laboratory Animal and Pathogen Bank, National Institute of Infectious Diseases, Tokyo, Japan
| | - Douglas W. Lowman
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Toshihiko Sugiki
- Laboratory of Molecular Biophysics, Institute for Protein Research, Osaka University, Osaka, Japan
| | - David L. Williams
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Sho Yamasaki
- Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, Japan
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, Japan
- Division of Molecular Design, Research Center for Systems Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
5
|
Cai R, Gong X, Li X, Jiang Y, Deng S, Tang J, Ge H, Wu C, Tang H, Wang G, Xie L, Chen X, Hu X, Feng J. Dectin-1 aggravates neutrophil inflammation through caspase-11/4-mediated macrophage pyroptosis in asthma. Respir Res 2024; 25:119. [PMID: 38459541 PMCID: PMC10921740 DOI: 10.1186/s12931-024-02743-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/20/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND The pattern recognition receptor Dectin-1 was initially discovered to play a pivotal role in mediating pulmonary antifungal immunity and promoting neutrophil-driven inflammation. Recent studies have revealed that Dectin-1 is overexpressed in asthma, but the specific mechanism remains elusive. Additionally, Dectin-1 has been implicated in promoting pyroptosis, a hallmark of severe asthma airway inflammation. Nevertheless, the involvement of the non-classical pyroptosis signal caspase-11/4 and its upstream regulatory mechanisms in asthma has not been completely explored. METHODS House dust mite (HDM)-induced mice was treated with Dectin-1 agonist Curdlan, Dectin-1 inhibitor Laminarin, and caspase-11 inhibitor wedelolactone separately. Subsequently, inflammatory cells in bronchoalveolar lavage fluid (BALF) were analyzed. Western blotting was performed to measure the protein expression of caspase-11 and gasdermin D (GSDMD). Cell pyroptosis and the expression of chemokine were detected in vitro. The correlation between Dectin-1 expression, pyroptosis factors and neutrophils in the induced sputum of asthma patients was analyzed. RESULTS Curdlan appeared to exacerbate neutrophil airway inflammation in asthmatic mice, whereas wedelolactone effectively alleviated airway inflammation aggravated by Curdlan. Moreover, Curdlan enhanced the release of caspase-11 activation fragments and N-terminal fragments of gasdermin D (GSDMD-N) stimulated by HDM both in vivo or in vitro. In mouse alveolar macrophages (MH-S cells), Curdlan/HDM stimulation resulted in vacuolar degeneration and elevated lactate dehydrogenase (LDH) release. In addition, there was an upregulation of neutrophil chemokines CXCL1, CXCL3, CXCL5 and their receptor CXCR2, which was suppressed by wedelolactone. In asthma patients, a positive correlation was observed between the expression of Dectin-1 on macrophages and caspase-4 (the human homology of caspase-11), and the proportion of neutrophils in induced sputum. CONCLUSION Dectin-1 activation in asthma induced caspase-11/4 mediated macrophage pyroptosis, which subsequently stimulated the secretion of chemokines, leading to the exacerbation of airway neutrophil inflammation.
Collapse
Grants
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 2022JJ30924 Natural Science Foundation of Hunan Province,China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 82270033 National Natural Science Foundation of China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
- 81873407 National Natural Science Foundation of China,China
Collapse
Affiliation(s)
- Runjin Cai
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoxiao Gong
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaozhao Li
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yuanyuan Jiang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shuanglinzi Deng
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jiale Tang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huan Ge
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Chendong Wu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huan Tang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Guo Wang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lei Xie
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xuemei Chen
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xinyue Hu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Juntao Feng
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
6
|
Generalov E, Yakovenko L. Receptor basis of biological activity of polysaccharides. Biophys Rev 2023; 15:1209-1222. [PMID: 37975017 PMCID: PMC10643635 DOI: 10.1007/s12551-023-01102-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/19/2023] [Indexed: 11/19/2023] Open
Abstract
Polysaccharides, the most diverse forms of organic molecules in nature, exhibit a large number of different biological activities, such as immunomodulatory, radioprotective, antioxidant, regenerative, metabolic, signaling, antitumor, and anticoagulant. The reaction of cells to a polysaccharide is determined by its specific interaction with receptors present on the cell surface, the type of cells, and their condition. The effect of many polysaccharides depends non-linearly on their concentration. The same polysaccharide in different conditions can have very different effects on cells and organisms, up to the opposite; therefore, when conducting studies of the biological activity of polysaccharides, both for the purpose of developing new drugs or approaches to the treatment of patients, and in order to clarify the features of intracellular processes, information about already known research results is needed. There is a lot of scattered data on the biological activities of polysaccharides, but there are few reviews that would consider natural polysaccharides from various sources and possible molecular mechanisms of their action. The purpose of this review is to present the main results published at different times in order to facilitate the search for information necessary for conducting relevant studies.
Collapse
Affiliation(s)
- Evgenii Generalov
- Faculty of Physics, M.V. Lomonosov Moscow State University, Moscow, 119991 Russia
| | - Leonid Yakovenko
- Faculty of Physics, M.V. Lomonosov Moscow State University, Moscow, 119991 Russia
| |
Collapse
|
7
|
Elbaset MA, Mohamed BMSA, Moustafa PE, Mansour DF, Afifi SM, Esatbeyoglu T, Abdelrahman SSM, Fayed HM. Erythropoietin Suppresses the Hepatic Fibrosis Caused by Thioacetamide: Role of the PI3K/Akt and TLR4 Signaling Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:5514248. [PMID: 37649466 PMCID: PMC10465256 DOI: 10.1155/2023/5514248] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/16/2023] [Accepted: 07/21/2023] [Indexed: 09/01/2023]
Abstract
Erythropoietin (EPO) is recognized for its function in erythropoiesis; however, its potential antifibrotic effect against liver fibrosis remains unknown. This study examined whether EPO affects thioacetamide (TAA)-induced liver fibrosis by concentrating on the Toll-like receptor 4 (TLR4) cascade and the phosphatidylinositol 3-kinase (PI3K)/Akt pathway as possible pathways. Male Wistar rats were randomized into four groups, which included: the negative control group, the TAA group (intraperitoneal; TAA 100 mg/kg three times per week for 2 weeks), and EPO-treated groups (150 and 300 IU/kg, i.p.) for 2 weeks after TAA injections. EPO attenuated hepatic fibrosis in a dosage-dependent way, as manifested by the diminution in serum alanine aminotransferase and aspartate aminotransferase activities, as well as the increase in albumin level. EPO inhibited the increase in tissue levels of tumor necrosis factors-α, interleukin-1β, transforming growth factor-β1, and TLR4 and raised tissue levels of PI3K and p-PI3K. EPO antioxidant properties were demonstrated by restoring hepatic glutathione and superoxide dismutase by preventing the accumulation of hepatic malondialdehyde. Further, EPO increased the protein expression of PI3K and Akt and decreased TLR4 protein expression. Immunohistochemically, EPO treatment altered tissue histology and downregulated mitogen-activated protein kinase protein expression. Overall, the research suggested that EPO could prevent TAA-induced hepatic fibrosis through upregulating the PI3K/Akt signaling cascade and downregulation the TLR4 downstream axis.
Collapse
Affiliation(s)
- Marawan A. Elbaset
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Bohouth St., Dokki, P.O. Box 12622, Cairo, Egypt
| | - Bassim M. S. A. Mohamed
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Bohouth St., Dokki, P.O. Box 12622, Cairo, Egypt
| | - Passant E. Moustafa
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Bohouth St., Dokki, P.O. Box 12622, Cairo, Egypt
| | - Dina F. Mansour
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Bohouth St., Dokki, P.O. Box 12622, Cairo, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Galala University, Attaka, Suez, Egypt
| | - Sherif M. Afifi
- Pharmacognosy Department, Faculty of Pharmacy, University of Sadat City, Sadat City 32897, Egypt
| | - Tuba Esatbeyoglu
- Department of Food Development and Food Quality, Institute of Food Science and Human Nutrition, Gottfried Wilhelm Leibniz University Hannover, Am Kleinen Felde 30, Hannover 30167, Germany
| | - Sahar S. M. Abdelrahman
- Department of Pathology, College of Veterinary Medicine, Cairo University, P.O. Box 12211, Cairo, Egypt
| | - Hany M. Fayed
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Bohouth St., Dokki, P.O. Box 12622, Cairo, Egypt
| |
Collapse
|
8
|
Yang N, Wang M, Lin K, Wang M, Xu D, Han X, Zhao X, Wang Y, Wu G, Luo W, Liang G, Shan P. Dectin-1 deficiency alleviates diabetic cardiomyopathy by attenuating macrophage-mediated inflammatory response. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166710. [PMID: 37054997 DOI: 10.1016/j.bbadis.2023.166710] [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: 02/08/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/15/2023]
Abstract
Cardiovascular diseases are the primary cause of mortality in patients with diabetes and obesity. Hyperglycemia and hyperlipidemia in diabetes alters cardiac function, which is associated with broader cellular processes such as aberrant inflammatory signaling. Recent studies have shown that a pattern recognition receptor called Dectin-1, expressed on macrophages, mediates pro-inflammatory responses in innate immunity. In the present study, we examined the role of Dectin-1 in the pathogenesis of diabetic cardiomyopathy. We observed increased Dectin-1 expression in heart tissues of diabetic mice and localized the source to macrophages. We then investigated the cardiac function in Dectin-1-deficient mice with STZ-induced type 1 diabetes and high-fat-diet-induced type 2 diabetes. Our results show that Dectin-1 deficient mice are protected against diabetes-induced cardiac dysfunction, cardiomyocyte hypertrophy, tissue fibrosis, and inflammation. Mechanistically, our studies show that Dectin-1 is important for cell activation and induction of inflammatory cytokines in high-concentration glucose and palmitate acid (HG + PA)-challenged macrophages. Deficiency of Dectin-1 generate fewer paracrine inflammatory factors capable of causing cardiomyocyte hypertrophy and fibrotic responses in cardiac fibroblasts. In conclusion, this study provides evidence that Dectin-1 mediates diabetes-induced cardiomyopathy through regulating inflammation. Dectin-1 may be a potential target to combat diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Na Yang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Minxiu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ke Lin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mengyang Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Diyun Xu
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xue Han
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xia Zhao
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gaojun Wu
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wu Luo
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guang Liang
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China; School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Peiren Shan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
9
|
Tang C, Sun H, Kadoki M, Han W, Ye X, Makusheva Y, Deng J, Feng B, Qiu D, Tan Y, Wang X, Guo Z, Huang C, Peng S, Chen M, Adachi Y, Ohno N, Trombetta S, Iwakura Y. Blocking Dectin-1 prevents colorectal tumorigenesis by suppressing prostaglandin E2 production in myeloid-derived suppressor cells and enhancing IL-22 binding protein expression. Nat Commun 2023; 14:1493. [PMID: 36932082 PMCID: PMC10023663 DOI: 10.1038/s41467-023-37229-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 03/07/2023] [Indexed: 03/19/2023] Open
Abstract
Dectin-1 (gene Clec7a), a receptor for β-glucans, plays important roles in the host defense against fungi and immune homeostasis of the intestine. Although this molecule is also suggested to be involved in the regulation of tumorigenesis, the role in intestinal tumor development remains to be elucidated. In this study, we find that azoxymethane-dextran-sodium-sulfate-induced and ApcMin-induced intestinal tumorigenesis are suppressed in Clec7a-/- mice independently from commensal microbiota. Dectin-1 is preferentially expressed on myeloid-derived suppressor cells (MDSCs). In the Clec7a-/- mouse colon, the proportion of MDSCs and MDSC-derived prostaglandin E2 (PGE2) levels are reduced, while the expression of IL-22 binding protein (IL-22BP; gene Il22ra2) is upregulated. Dectin-1 signaling induces PGE2-synthesizing enzymes and PGE2 suppresses Il22ra2 expression in vitro and in vivo. Administration of short chain β-glucan laminarin, an antagonist of Dectin-1, suppresses the development of mouse colorectal tumors. Furthermore, in patients with colorectal cancer (CRC), the expression of CLEC7A is also observed in MDSCs and correlated with the death rate and tumor severity. Dectin-1 signaling upregulates PGE2-synthesizing enzyme expression and PGE2 suppresses IL22RA2 expression in human CRC-infiltrating cells. These observations indicate a role of the Dectin-1-PGE2-IL-22BP axis in regulating intestinal tumorigenesis, suggesting Dectin-1 as a potential target for CRC therapy.
Collapse
Affiliation(s)
- Ce Tang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China.
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China.
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan.
| | - Haiyang Sun
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan
| | - Motohiko Kadoki
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan
| | - Wei Han
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan
| | - Xiaoqi Ye
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan
| | - Yulia Makusheva
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan
| | - Jianping Deng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Bingbing Feng
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Ding Qiu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Ying Tan
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Xinying Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Zehao Guo
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Chanyan Huang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Sui Peng
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Minhu Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital, Sun Yat-sen University, No.58, Zhong Shan Er Lu, 510080, Guangzhou, Guangdong Province, China
| | - Yoshiyuki Adachi
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Naohito Ohno
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan
| | - Sergio Trombetta
- Boehringer Ingelheim USA, 900 Ridgebury Rd, Ridgefield, CT, 06877, USA
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Yamazaki 2669, Noda-shi, Chiba, 278-0022, Japan.
| |
Collapse
|
10
|
Hatinguais R, Willment JA, Brown GD. C-type lectin receptors in antifungal immunity: Current knowledge and future developments. Parasite Immunol 2023; 45:e12951. [PMID: 36114607 PMCID: PMC10078331 DOI: 10.1111/pim.12951] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 09/05/2022] [Accepted: 09/12/2022] [Indexed: 01/31/2023]
Abstract
C-type lectin receptors (CLRs) constitute a category of innate immune receptors that play an essential role in the antifungal immune response. For over two decades, scientists have uncovered what are the fungal ligands recognized by CLRs and how these receptors initiate the immune response. Such studies have allowed the identification of genetic polymorphisms in genes encoding for CLRs or for proteins involved in the signalisation cascade they trigger. Nevertheless, our understanding of how these receptors functions and the full extent of their function during the antifungal immune response is still at its infancy. In this review, we summarize some of the main findings about CLRs in antifungal immunity and discuss what the future might hold for the field.
Collapse
Affiliation(s)
- Remi Hatinguais
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Janet A Willment
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Gordon D Brown
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| |
Collapse
|
11
|
Wang MX, Luo W, Ye L, Jin LM, Yang B, Zhang QH, Qian JC, Wang Y, Zhang Y, Liang G. Dectin-1 plays a deleterious role in high fat diet-induced NAFLD of mice through enhancing macrophage activation. Acta Pharmacol Sin 2023; 44:120-132. [PMID: 35689091 DOI: 10.1038/s41401-022-00926-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/24/2022] [Indexed: 01/18/2023] Open
Abstract
The innate immune response and inflammation contribute to hepatic steatosis and non-alcoholic fatty liver disease (NAFLD). Dectin-1 is a pathogen recognition receptor in innate immunity. In this study, we investigated the role of Dectin-1 in the pathogenesis of NAFLD. We first showed that Dectin-1 expression was significantly elevated in liver tissues of patients with NASH. NAFLD was induced in mice by feeding high fat diet (HFD) for 24 weeks. At the end of treatment, mice were sacrificed, and their blood and liver tissues were collected for analyses. We showed HFD feeding also increased liver Dectin-1 levels in mice, associated with macrophage infiltration. Either gene knockout or co-administration of a Dectin-1 antagonist laminarin (150 mg/kg twice a day, ip, from 16th week to 24th week) largely protected the livers from HFD-induced lipid accumulation, fibrosis, and elaboration of inflammatory responses. In primary mouse peritoneal macrophages (MPMs), challenge with palmitate (PA, 200 μM), an abundant saturated fatty acid found in NAFLD, significantly activated Dectin-1 signaling pathway, followed by transcriptionally regulated production of pro-inflammatory cytokines. Dectin-1 was required for hepatic macrophage activation and inflammatory factor induction. Condition media generated from Dectin-1 deficient macrophages failed to cause hepatocyte lipid accumulation and hepatic stellate activation. In conclusion, this study provides the primary evidence supporting a deleterious role for Dectin-1 in NAFLD through enhancing macrophage pro-inflammatory responses and suggests that it can be targeted to prevent inflammatory NAFLD.
Collapse
Affiliation(s)
- Min-Xiu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.,Medical Research Center, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Lin Ye
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Lei-Ming Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Bin Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Qian-Hui Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jian-Chang Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yi Zhang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China. .,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 311399, China.
| |
Collapse
|
12
|
Xiang C, Li H, Tang W. Targeting CSF-1R represents an effective strategy in modulating inflammatory diseases. Pharmacol Res 2023; 187:106566. [PMID: 36423789 DOI: 10.1016/j.phrs.2022.106566] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/12/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
Colony-stimulating factor-1 receptor (CSF-1R), also known as FMS kinase, is a type I single transmembrane protein mainly expressed in myeloid cells, such as monocytes, macrophages, glial cells, and osteoclasts. The endogenous ligands, colony-stimulating factor-1 (CSF-1) and Interleukin-34 (IL-34), activate CSF-1R and downstream signaling pathways including PI3K-AKT, JAK-STATs, and MAPKs, and modulate the proliferation, differentiation, migration, and activation of target immune cells. Over the past decades, the promising therapeutic potential of CSF-1R signaling inhibition has been widely studied for decreasing immune suppression and escape in tumors, owing to depletion and reprogramming of tumor-associated macrophages. In addition, the excessive activation of CSF-1R in inflammatory diseases is consecutively uncovered in recent years, which may result in inflammation in bone, kidney, lung, liver and central nervous system. Agents against CSF-1R signaling have been increasingly investigated in preclinical or clinical studies for inflammatory diseases treatment. However, the pathological mechanism of CSF-1R in inflammation is indistinct and whether CSF-1R signaling can be identified as biomarkers remains controversial. With the background information aforementioned, this review focus on the dialectical roles of CSF-1R and its ligands in regulating innate immune cells and highlights various therapeutic implications of blocking CSF-1R signaling in inflammatory diseases.
Collapse
Affiliation(s)
- Caigui Xiang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heng Li
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Wei Tang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
13
|
Candida Administration in 5/6 Nephrectomized Mice Enhanced Fibrosis in Internal Organs: An Impact of Lipopolysaccharide and (1→3)-β-D-Glucan from Leaky Gut. Int J Mol Sci 2022; 23:ijms232415987. [PMID: 36555628 PMCID: PMC9784901 DOI: 10.3390/ijms232415987] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Uremic toxins and gut dysbiosis in advanced chronic kidney disease (CKD) can induce gut leakage, causing the translocation of gut microbial molecules into the systemic circulation. Lipopolysaccharide (LPS) and (1→3)-β-D-glucan (BG) are the major gut microbial molecules of Gram-negative bacteria and fungi, respectively, and can induce inflammation in several organs. Here, the fibrosis in the kidney, liver, and heart was investigated in oral C. albicans-administered 5/6 nephrectomized (Candida-5/6 Nx) mice. At 20 weeks post 5/6 Nx, Candida-5/6 Nx mice demonstrated increased 24 h proteinuria, liver enzymes, and serum cytokines (TNF-α, IL-6, and IL-10), but not weight loss, systolic blood pressure, hematocrit, serum creatinine, or gut-derived uremic toxins (TMAO and indoxyl sulfate), compared to in 5/6 Nx alone. The gut leakage in Candida-5/6 Nx was more severe, as indicated by FITC-dextran assay, endotoxemia, and serum BG. The areas of fibrosis from histopathology, along with the upregulated gene expression of Toll-like receptor 4 (TLR-4) and Dectin-1, the receptors for LPS and BG, respectively, were higher in the kidney, liver, and heart. In vitro, LPS combined with BG increased the supernatant IL-6 and TNF-α, upregulated the genes of pro-inflammation and pro-fibrotic processes, Dectin-1, and TLR-4 in renal tubular (HK-2) cells and hepatocytes (HepG2), when compared with LPS or BG alone. This supported the pro-inflammation-induced fibrosis and the possible LPS-BG additive effects on kidney and liver fibrosis. In conclusion, uremia-induced leaky gut causes the translocation of gut LPS and BG into circulation, which activates the pro-inflammatory and pro-fibrotic pathways, causing internal organ fibrosis. Our results support the crosstalk among several organs in CKD through a leaky gut.
Collapse
|
14
|
Qu W, Qiao S, Liu L, Chen Y, Peng C, Hou Y, Xu Z, Lv M, Wang T. Dectin3 protects against hepatocellular carcinoma by regulating glycolysis of macrophages. Int Immunopharmacol 2022; 113:109384. [DOI: 10.1016/j.intimp.2022.109384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
|
15
|
Zhao Q, Bai J, Chen Y, Liu X, Zhao S, Ling G, Jia S, Zhai F, Xiang R. An optimized herbal combination for the treatment of liver fibrosis: Hub genes, bioactive ingredients, and molecular mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115567. [PMID: 35870684 DOI: 10.1016/j.jep.2022.115567] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/30/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liver fibrosis is a chronic liver disease that can lead to cirrhosis, liver failure, and hepatocellular carcinoma, and it is associated with long-term adverse outcomes and mortality. As a primary resource for complementary and alternative medicine, traditional Chinese medicine (TCM) has accumulated a large number of effective formulas for the treatment of liver fibrosis in clinical practice. However, studies on how to systematically optimize TCM formulas are still lacking. AIM OF THE REVIEW To provide a methodological reference for the systematic optimization of TCM formulae against liver fibrosis and explored the underlying molecular mechanisms; To provide an efficient method for searching for lead compounds from natural sources and developing from herbal medicines; To enable clinicians and patients to make more reasonable choices and promote the effective treatment toward those patients with liver fibrosis. MATERIALS AND METHODS TCM formulas related to treating liver fibrosis were collected from the Web of Science, PubMed, the China National Knowledge Infrastructure (CNKI), Wan Fang, and the Chinese Scientific Journals Database (VIP). Furthermore, the TCM compatibility patterns were mined using association analysis. The core TCM combinations were found by designing an optimized formulas algorithm. Finally, the hub target proteins, potential molecular mechanisms, and active compounds were explored through integrative pharmacology and docking-based inverse virtual screening (IVS) approaches. RESULTS We found that the herbs for reinforcing deficiency, activating blood, removing blood stasis, and clearing heat were the basis of TCM formulae patterns. Furthermore, the combination of Salviae Miltiorrhizae (Salvia miltiorrhiza Bunge; Chinese salvia/Danshen), Astragali Radix (Astragalus membranaceus (Fisch.) Bunge; Astragalus/Huangqi), and Radix Bupleuri (Bupleurum chinense DC.; Bupleurum/Chaihu) was identified as core groups. A total of six targets (TNF, STAT3, EGFR, IL2, ICAM1, PTGS2) play a pivotal role in TCM-mediated liver fibrosis inhibition. (-)-Cryptotanshinone, Tanshinaldehyde, Ononin, Thymol, Daidzein, and Formononetin were identified as active compounds in TCM. And mechanistically, TCM could affect the development of liver fibrosis by regulating inflammation, immunity, angiogenesis, antioxidants, and involvement in TNF, MicroRNAs, Jak-STAT, NF-kappa B, and C-type lectin receptors (CLRs) signaling pathways. Molecular docking results showed that key components had good potential to bind to the target genes. CONCLUSION In summary, this study provides a methodological reference for the systematic optimization of TCM formulae and exploration of underlying molecular mechanisms.
Collapse
Affiliation(s)
- Qianqian Zhao
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Jinwei Bai
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Yiwei Chen
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Xin Liu
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Shangfeng Zhao
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Guixia Ling
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Shubing Jia
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Fei Zhai
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Rongwu Xiang
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China; Liaoning Professional Technology Innovation Center on Medical Big Data and Artificial Intelligence, Shenyang, 110016, China.
| |
Collapse
|
16
|
Zhang L, Chai D, Chen C, Li C, Qiu Z, Kuang T, Parveena M, Dong K, Yu J, Deng W, Wang W. Mycobiota and C-Type Lectin Receptors in Cancers: Know thy Neighbors. Front Microbiol 2022; 13:946995. [PMID: 35910636 PMCID: PMC9326027 DOI: 10.3389/fmicb.2022.946995] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 11/15/2022] Open
Abstract
Numerous studies have demonstrated the importance of gut bacteria in the development of malignancy, while relatively little research has been done on gut mycobiota. As a part of the gut microbiome, the percentage of gut mycobiota is negligible compared to gut bacteria. However, the effect of gut fungi on human health and disease is significant. This review systematically summarizes the research progress on mycobiota, especially gut fungi, in patients with head and neck cancer (HNC), esophageal cancer (EC), gastric cancer (GC), colorectal cancer (CRC), hepatocellular carcinoma (HCC), pancreatic cancer, melanoma, breast cancer, and lung carcinoma-induced cachexia. Moreover, we also describe, for the first time in detail, the role of the fungal recognition receptors, C-type lectin receptors (CLRs) (Dectin-1, Dectin-2, Dectin-3, and Mincle) and their downstream effector caspase recruitment domain-containing protein 9 (CARD9), in tumors to provide a reference for further research on intestinal fungi in the diagnosis and treatment of malignant tumors.
Collapse
Affiliation(s)
- Lilong Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Dongqi Chai
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Chen Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chunlei Li
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Zhendong Qiu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Tianrui Kuang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Mungur Parveena
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Keshuai Dong
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jia Yu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenhong Deng
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Wenhong Deng,
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Weixing Wang,
| |
Collapse
|
17
|
Zheng P, Zhang H, Jiang W, Wang L, Liu L, Zhou Y, Zhou L, Liu H. Establishment of a Prognostic Model of Lung Adenocarcinoma Based on Tumor Heterogeneity. Front Mol Biosci 2022; 9:807497. [PMID: 35480896 PMCID: PMC9035852 DOI: 10.3389/fmolb.2022.807497] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is one of the main cancer types due to its persistently high incidence and mortality, yet a simple and effective prognostic model is still lacking. This study aimed to identify independent prognostic genes related to the heterogeneity of lung adenocarcinoma (LUAD), generate a prognostic risk score model, and construct a nomogram in combination with other pathological characteristics to predict patients’ overall survival (OS). A significant amount of data pertaining to single-cell RNA sequencing (scRNA-seq), RNA sequencing (RNA-seq), and somatic mutation were used for data mining. After statistical analyses, a risk scoring model was established based on eight independent prognostic genes, and the OS of high-risk patients was significantly lower than that of low-risk patients. Interestingly, high-risk patients were more sensitive and effective to immune checkpoint blocking therapy. In addition, it was noteworthy that CCL20 not only affected prognosis and differentiation of LUAD but also led to poor histologic grade of tumor cells. Ultimately, combining risk score, clinicopathological information, and CCL20 mutation status, a nomogram with good predictive performance and high accuracy was established. In short, our research established a prognostic model that could be used to guide clinical practice based on the constantly updated big multi-omics data. Finally, this analysis revealed that CCL20 may become a potential therapeutic target for LUAD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ling Zhou
- *Correspondence: Ling Zhou, ; Huiguo Liu,
| | - Huiguo Liu
- *Correspondence: Ling Zhou, ; Huiguo Liu,
| |
Collapse
|
18
|
Keshvari S, Genz B, Teakle N, Caruso M, Cestari MF, Patkar OL, Tse BWC, Sokolowski KA, Ebersbach H, Jascur J, MacDonald KPA, Miller G, Ramm GA, Pettit AR, Clouston AD, Powell EE, Hume DA, Irvine KM. Therapeutic potential of macrophage colony-stimulating factor (CSF1) in chronic liver disease. Dis Model Mech 2022; 15:274391. [PMID: 35169835 PMCID: PMC9044210 DOI: 10.1242/dmm.049387] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/08/2022] [Indexed: 11/20/2022] Open
Abstract
Resident and recruited macrophages control the development and proliferation of the liver. We showed previously in multiple species that treatment with a macrophage colony stimulating factor (CSF1)-Fc fusion protein initiated hepatocyte proliferation and promoted repair in models of acute hepatic injury in mice. Here we investigated the impact of CSF1-Fc on resolution of advanced fibrosis and liver regeneration, utilizing a non-resolving toxin-induced model of chronic liver injury and fibrosis in C57BL/6J mice. Co-administration of CSF1-Fc with exposure to thioacetamide (TAA) exacerbated inflammation consistent with monocyte contributions to initiation of pathology. After removal of TAA, either acute or chronic CSF1-Fc treatment promoted liver growth, prevented progression and promoted resolution of fibrosis. Acute CSF1-Fc treatment was also anti-fibrotic and pro-regenerative in a model of partial hepatectomy in mice with established fibrosis. The beneficial impacts of CSF1-Fc treatment were associated with monocyte-macrophage recruitment and increased expression of remodeling enzymes and growth factors. These studies indicate that CSF1-dependent macrophages contribute to both initiation and resolution of fibrotic injury and that CSF1-Fc has therapeutic potential in human liver disease.
Collapse
Affiliation(s)
- Sahar Keshvari
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Berit Genz
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Ngari Teakle
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Melanie Caruso
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Michelle F Cestari
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Omkar L Patkar
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Brian W C Tse
- Preclinical Imaging Facility, Translational Research Institute, Brisbane, Queensland, Australia
| | - Kamil A Sokolowski
- Preclinical Imaging Facility, Translational Research Institute, Brisbane, Queensland, Australia
| | - Hilmar Ebersbach
- Novartis Institutes for Biomedical Research (NIBR), Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Julia Jascur
- Novartis Institutes for Biomedical Research (NIBR), Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | | | | | - Grant A Ramm
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Allison R Pettit
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Andrew D Clouston
- Envoi Specialist Pathologists, Brisbane, Qld, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Elizabeth E Powell
- Faculty of Medicine, The University of Queensland, Brisbane, Australia.,Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - David A Hume
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Katharine M Irvine
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
19
|
Patten DA, Wilkinson AL, O'Keeffe A, Shetty S. Scavenger Receptors: Novel Roles in the Pathogenesis of Liver Inflammation and Cancer. Semin Liver Dis 2022; 42:61-76. [PMID: 34553345 PMCID: PMC8893982 DOI: 10.1055/s-0041-1733876] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The scavenger receptor superfamily represents a highly diverse collection of evolutionarily-conserved receptors which are known to play key roles in host homeostasis, the most prominent of which is the clearance of unwanted endogenous macromolecules, such as oxidized low-density lipoproteins, from the systemic circulation. Members of this family have also been well characterized in their binding and internalization of a vast range of exogenous antigens and, consequently, are generally considered to be pattern recognition receptors, thus contributing to innate immunity. Several studies have implicated scavenger receptors in the pathophysiology of several inflammatory diseases, such as Alzheimer's and atherosclerosis. Hepatic resident cellular populations express a diverse complement of scavenger receptors in keeping with the liver's homeostatic functions, but there is gathering interest in the contribution of these receptors to hepatic inflammation and its complications. Here, we review the expression of scavenger receptors in the liver, their functionality in liver homeostasis, and their role in inflammatory liver disease and cancer.
Collapse
Affiliation(s)
- Daniel A. Patten
- National Institute for Health Research Birmingham Liver Biomedical Research Unit, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Alex L. Wilkinson
- National Institute for Health Research Birmingham Liver Biomedical Research Unit, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Ayla O'Keeffe
- National Institute for Health Research Birmingham Liver Biomedical Research Unit, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Shishir Shetty
- National Institute for Health Research Birmingham Liver Biomedical Research Unit, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
20
|
Goedhart M, Slot E, Pascutti MF, Geerman S, Rademakers T, Nota B, Huveneers S, van Buul JD, MacNamara KC, Voermans C, Nolte MA. Bone Marrow Harbors a Unique Population of Dendritic Cells with the Potential to Boost Neutrophil Formation upon Exposure to Fungal Antigen. Cells 2021; 11:55. [PMID: 35011617 PMCID: PMC8750392 DOI: 10.3390/cells11010055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 02/01/2023] Open
Abstract
Apart from controlling hematopoiesis, the bone marrow (BM) also serves as a secondary lymphoid organ, as it can induce naïve T cell priming by resident dendritic cells (DC). When analyzing DCs in murine BM, we uncovered that they are localized around sinusoids, can (cross)-present antigens, become activated upon intravenous LPS-injection, and for the most part belong to the cDC2 subtype which is associated with Th2/Th17 immunity. Gene-expression profiling revealed that BM-resident DCs are enriched for several c-type lectins, including Dectin-1, which can bind beta-glucans expressed on fungi and yeast. Indeed, DCs in BM were much more efficient in phagocytosis of both yeast-derived zymosan-particles and Aspergillus conidiae than their splenic counterparts, which was highly dependent on Dectin-1. DCs in human BM could also phagocytose zymosan, which was dependent on β1-integrins. Moreover, zymosan-stimulated BM-resident DCs enhanced the differentiation of hematopoietic stem and progenitor cells towards neutrophils, while also boosting the maintenance of these progenitors. Our findings signify an important role for BM DCs as translators between infection and hematopoiesis, particularly in anti-fungal immunity. The ability of BM-resident DCs to boost neutrophil formation is relevant from a clinical perspective and contributes to our understanding of the increased susceptibility for fungal infections following BM damage.
Collapse
Affiliation(s)
- Marieke Goedhart
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
| | - Edith Slot
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
| | - Maria F. Pascutti
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
| | - Sulima Geerman
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
| | - Timo Rademakers
- Molecular Cell Biology Lab, Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (T.R.); (S.H.); (J.D.v.B.)
| | - Benjamin Nota
- Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands;
| | - Stephan Huveneers
- Molecular Cell Biology Lab, Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (T.R.); (S.H.); (J.D.v.B.)
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jaap D. van Buul
- Molecular Cell Biology Lab, Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (T.R.); (S.H.); (J.D.v.B.)
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Katherine C. MacNamara
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208, USA;
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Martijn A. Nolte
- Department of Hematopoiesis, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands; (M.G.); (E.S.); (M.F.P.); (S.G.); (C.V.)
- Department of Molecular Hematology, Sanquin Research, Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands;
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
21
|
Cao X, Cordova AF, Li L. Therapeutic Interventions Targeting Innate Immune Receptors: A Balancing Act. Chem Rev 2021; 122:3414-3458. [PMID: 34870969 DOI: 10.1021/acs.chemrev.1c00716] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The innate immune system is an organism's first line of defense against an onslaught of internal and external threats. The downstream adaptive immune system has been a popular target for therapeutic intervention, while there is a relative paucity of therapeutics targeting the innate immune system. However, the innate immune system plays a critical role in many human diseases, such as microbial infection, cancer, and autoimmunity, highlighting the need for ongoing therapeutic research. In this review, we discuss the major innate immune pathways and detail the molecular strategies underpinning successful therapeutics targeting each pathway as well as previous and ongoing efforts. We will also discuss any recent discoveries that could inform the development of novel therapeutic strategies. As our understanding of the innate immune system continues to develop, we envision that therapies harnessing the power of the innate immune system will become the mainstay of treatment for a wide variety of human diseases.
Collapse
|
22
|
Zhang S, Li X, Tang C, Kuang W. Inflammation-Related Long Non-Coding RNA Signature Predicts the Prognosis of Gastric Carcinoma. Front Genet 2021; 12:736766. [PMID: 34819945 PMCID: PMC8607501 DOI: 10.3389/fgene.2021.736766] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/05/2021] [Indexed: 12/25/2022] Open
Abstract
Background: Gastric carcinoma (GC) is a molecularly and phenotypically highly heterogeneous disease, making the prognostic prediction challenging. On the other hand, Inflammation as part of the active cross-talk between the tumor and the host in the tumor or its microenvironment could affect prognosis. Method: We established a prognostic multi lncRNAs signature that could better predict the prognosis of GC patients based on inflammation-related differentially expressed lncRNAs in GC. Results: We identified 10 differently expressed lncRNAs related to inflammation associated with GC prognosis. Kaplan-Meier survival analysis demonstrated that high-risk inflammation-related lncRNAs signature was related to poor prognosis of GC. Moreover, the inflammation-related lncRNAs signature had an AUC of 0.788, proving their utility in predicting GC prognosis. Indeed, our risk signature is more precise in predicting the prognosis of GC patients than traditional clinicopathological manifestations. Immune and tumor-related pathways for individuals in the low and high-risk groups were further revealed by GSEA. Moreover, TCGA based analysis revealed significant differences in HLA, MHC class-I, cytolytic activity, parainflammation, co-stimulation of APC, type II INF response, and type I INF response between the two risk groups. Immune checkpoints revealed CD86, TNFSF18, CD200, and LAIR1 were differently expressed between lowand high-risk groups. Conclusion: A novel inflammation-related lncRNAs (AC015660.1, LINC01094, AL512506.1, AC124067.2, AC016737.1, AL136115.1, AP000695.1, AC104695.3, LINC00449, AC090772.1) signature may provide insight into the new therapies and prognosis prediction for GC patients.
Collapse
Affiliation(s)
- ShuQiao Zhang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - XinYu Li
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - ChunZhi Tang
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - WeiHong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
23
|
Bruneau A, Hundertmark J, Guillot A, Tacke F. Molecular and Cellular Mediators of the Gut-Liver Axis in the Progression of Liver Diseases. Front Med (Lausanne) 2021; 8:725390. [PMID: 34650994 PMCID: PMC8505679 DOI: 10.3389/fmed.2021.725390] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022] Open
Abstract
The gut-liver axis covers the bidirectional communication between the gut and the liver, and thus includes signals from liver-to-gut (e.g., bile acids, immunoglobulins) and from gut-to-liver (e.g., nutrients, microbiota-derived products, and recirculating bile acids). In a healthy individual, liver homeostasis is tightly controlled by the mostly tolerogenic liver resident macrophages, the Kupffer cells, capturing the gut-derived antigens from the blood circulation. However, disturbances of the gut-liver axis have been associated to the progression of varying chronic liver diseases, such as non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, and primary sclerosing cholangitis. Notably, changes of the gut microbiome, or intestinal dysbiosis, combined with increased intestinal permeability, leads to the translocation of gut-derived bacteria or their metabolites into the portal vein. In the context of concomitant or subsequent liver inflammation, the liver is then infiltrated by responsive immune cells (e.g., monocytes, neutrophils, lymphoid, or dendritic cells), and microbiota-derived products may provoke or exacerbate innate immune responses, hence perpetuating liver inflammation and fibrosis, and potentiating the risks of developing cirrhosis. Similarly, food derived antigens, bile acids, danger-, and pathogen-associated molecular patterns are able to reshape the liver immune microenvironment. Immune cell intracellular signaling components, such as inflammasome activation, toll-like receptor or nucleotide-binding oligomerization domain-like receptors signaling, are potent targets of interest for the modulation of the immune response. This review describes the current understanding of the cellular landscape and molecular pathways involved in the gut-liver axis and implicated in chronic liver disease progression. We also provide an overview of innovative therapeutic approaches and current clinical trials aiming at targeting the gut-liver axis for the treatment of patients with chronic liver and/or intestinal diseases.
Collapse
Affiliation(s)
- Alix Bruneau
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Jana Hundertmark
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| |
Collapse
|
24
|
Epoxyeicosatrienoic Acids and Fibrosis: Recent Insights for the Novel Therapeutic Strategies. Int J Mol Sci 2021; 22:ijms221910714. [PMID: 34639055 PMCID: PMC8509622 DOI: 10.3390/ijms221910714] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 12/27/2022] Open
Abstract
Organ fibrosis often ends in eventual organ failure and leads to high mortality. Although researchers have identified many effector cells and molecular pathways, there are few effective therapies for fibrosis to date and the underlying mechanism needs to be examined and defined further. Epoxyeicosatrienoic acids (EETs) are endogenous lipid metabolites of arachidonic acid (ARA) synthesized by cytochrome P450 (CYP) epoxygenases. EETs are rapidly metabolized primarily via the soluble epoxide hydrolase (sEH) pathway. The sEH pathway produces dihydroxyeicosatrienoic acids (DHETs), which have lower activity. Stabilized or increased EETs levels exert several protective effects, including pro-angiogenesis, anti-inflammation, anti-apoptosis, and anti-senescence. Currently, intensive investigations are being carried out on their anti-fibrotic effects in the kidney, heart, lung, and liver. The present review provides an update on how the stabilized or increased production of EETs is a reasonable theoretical basis for fibrosis treatment.
Collapse
|
25
|
Zhuang T, Li W, Yang L, Wang Z, Ding L, Zhou M. Gut Microbiota: Novel Therapeutic Target of Ginsenosides for the Treatment of Obesity and Its Complications. Front Pharmacol 2021; 12:731288. [PMID: 34512356 PMCID: PMC8429618 DOI: 10.3389/fphar.2021.731288] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity, generally characterized by excessive lipid accumulation, is a metabolic threat worldwide due to its rapid growth in global prevalence. Ginsenosides are crucial components derived from natural plants that can confer metabolic benefits for obese patients. Considering the low bioavailability and degradable properties of ginsenosides in vivo, it should be admitted that the mechanism of ginsenosides on anti-obesity contribution is still obscure. Recently, studies have indicated that ginsenoside intervention has beneficial metabolic effects on obesity and its complications because it allows for the correction of gut microbiota dysbiosis and regulates the secretion of related endogenous metabolites. In this review, we summarize the role of gut microbiota in the pathogenetic process of obesity, and explore the mechanism of ginsenosides for ameliorating obesity, which can modulate the composition of gut microbiota by improving the metabolism of intestinal endogenous substances and alleviating the level of inflammation. Ginsenosides are expected to become a promising anti-obesity medical intervention in the foreseeable clinical settings.
Collapse
Affiliation(s)
- Tongxi Zhuang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Center for Chinese Medicine Therapy and Systems Biology, Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Li
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Li Yang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Zhengtao Wang
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Lili Ding
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai, China
| | - Mingmei Zhou
- Center for Chinese Medicine Therapy and Systems Biology, Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Di Lorenzo F, Duda KA, Lanzetta R, Silipo A, De Castro C, Molinaro A. A Journey from Structure to Function of Bacterial Lipopolysaccharides. Chem Rev 2021; 122:15767-15821. [PMID: 34286971 DOI: 10.1021/acs.chemrev.0c01321] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Lipopolysaccharide (LPS) is a crucial constituent of the outer membrane of most Gram-negative bacteria, playing a fundamental role in the protection of bacteria from environmental stress factors, in drug resistance, in pathogenesis, and in symbiosis. During the last decades, LPS has been thoroughly dissected, and massive information on this fascinating biomolecule is now available. In this Review, we will give the reader a third millennium update of the current knowledge of LPS with key information on the inherent peculiar carbohydrate chemistry due to often puzzling sugar residues that are uniquely found on it. Then, we will drive the reader through the complex and multifarious immunological outcomes that any given LPS can raise, which is strictly dependent on its chemical structure. Further, we will argue about issues that still remain unresolved and that would represent the immediate future of LPS research. It is critical to address these points to complete our notions on LPS chemistry, functions, and roles, in turn leading to innovative ways to manipulate the processes involving such a still controversial and intriguing biomolecule.
Collapse
Affiliation(s)
- Flaviana Di Lorenzo
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy.,Task Force on Microbiome Studies, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy
| | - Katarzyna A Duda
- Research Center Borstel Leibniz Lung Center, Parkallee 4a, 23845 Borstel, Germany
| | - Rosa Lanzetta
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy
| | - Alba Silipo
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy.,Task Force on Microbiome Studies, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy
| | - Cristina De Castro
- Task Force on Microbiome Studies, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy.,Department of Agricultural Sciences, University of Naples Federico II, Via Università 96, 80055 Portici, Naples, Italy
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy.,Task Force on Microbiome Studies, University of Naples Federico II, Via Cinthia 4, 80126 Naples, Italy.,Department of Chemistry, School of Science, Osaka University, 1-1 Osaka University Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
27
|
Cui ZY, Wang G, Zhang J, Song J, Jiang YC, Dou JY, Lian LH, Nan JX, Wu YL. Parthenolide, bioactive compound of Chrysanthemum parthenium L., ameliorates fibrogenesis and inflammation in hepatic fibrosis via regulating the crosstalk of TLR4 and STAT3 signaling pathway. Phytother Res 2021; 35:5680-5693. [PMID: 34250656 DOI: 10.1002/ptr.7214] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 05/21/2021] [Accepted: 06/19/2021] [Indexed: 01/05/2023]
Abstract
The current study focused on the regulatory effects of parthenolide (PNL), a bioactive component derived from Chrysanthemum parthenium L., against hepatic fibrosis via regulating the crosstalk of toll-like receptor 4 (TLR4) and signal transducer and activator of transcription 3 (STAT3) in activated hepatic stellate cells (HSCs). HSCs or Raw 264.7 macrophages were activated by TGF-β or LPS for 1 hr, respectively, and then treated with PNL, CLI-095 (TLR4 inhibitor), or Niclosamide (STAT3 inhibitor) for the indicated time to detect the crosstalk of TLR4 and STAT3. PNL significantly decreased the expressions of α-SMA, collagen I, and the ratio of TIMP1 and MMP13 in TGF-β-activated HSCs. PNL significantly reduced the releases of pro-inflammatory cytokines, including IL-6, IL-1β, IL-1α, IL-18, and regulated signaling P2X7r/NLRP3 axis activation. PNL obviously induced the apoptosis of activated HSCs by regulating bcl-2 and caspases family. PNL significantly inhibited the expressions of TLR4 and STAT3, including their downstream signaling. PNL could regulate the crosstalk of TLR4 and STAT3, which were verified by their inhibitors in activated HSCs or Raw 264.7 cell macrophages. Thus, PNL could decrease the expressions of fibrosis markers, reduce the releases of inflammatory cytokines, and also induce the apoptosis of activated HSCs. In conclusion, PNL could bi-directionally inhibit TLR4 and STAT3 signaling pathway, suggesting that blocking the crosstalk of TLR4 and STAT3 might be the potential mechanism of PNL against hepatic fibrosis.
Collapse
Affiliation(s)
- Zhen-Yu Cui
- Key Laboratory for Traditional Chinese Korean Medicine of Jilin Province, College of Pharmacy, Yanbian University, Yanji, China
| | - Ge Wang
- Key Laboratory for Traditional Chinese Korean Medicine of Jilin Province, College of Pharmacy, Yanbian University, Yanji, China
| | - Jing Zhang
- Research and Development Center, Liaoning Shengjing Stem cell technology Co., Ltd, Shenyang, China
| | - Jian Song
- Key Laboratory for Traditional Chinese Korean Medicine of Jilin Province, College of Pharmacy, Yanbian University, Yanji, China
| | - Yu-Chen Jiang
- Key Laboratory for Traditional Chinese Korean Medicine of Jilin Province, College of Pharmacy, Yanbian University, Yanji, China
| | - Jia-Yi Dou
- Key Laboratory for Traditional Chinese Korean Medicine of Jilin Province, College of Pharmacy, Yanbian University, Yanji, China
| | - Li-Hua Lian
- Key Laboratory for Traditional Chinese Korean Medicine of Jilin Province, College of Pharmacy, Yanbian University, Yanji, China
| | - Ji-Xing Nan
- Key Laboratory for Traditional Chinese Korean Medicine of Jilin Province, College of Pharmacy, Yanbian University, Yanji, China.,Clinical Research Centre, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yan-Ling Wu
- Key Laboratory for Traditional Chinese Korean Medicine of Jilin Province, College of Pharmacy, Yanbian University, Yanji, China
| |
Collapse
|
28
|
Wan J, Weiss E, Ben Mkaddem S, Mabire M, Choinier PM, Picq O, Thibault-Sogorb T, Hegde P, Pishvaie D, Bens M, Broer L, Gilgenkrantz H, Moreau R, Saveanu L, Codogno P, Monteiro RC, Lotersztajn S. LC3-associated phagocytosis protects against inflammation and liver fibrosis via immunoreceptor inhibitory signaling. Sci Transl Med 2021; 12:12/539/eaaw8523. [PMID: 32295902 DOI: 10.1126/scitranslmed.aaw8523] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 10/11/2019] [Accepted: 02/09/2020] [Indexed: 12/15/2022]
Abstract
Sustained hepatic and systemic inflammation, particularly originating from monocytes/macrophages, is a driving force for fibrosis progression to end-stage cirrhosis and underlies the development of multiorgan failure. Reprogramming monocyte/macrophage phenotype has emerged as a strategy to limit inflammation during chronic liver injury. Here, we report that LC3-associated phagocytosis (LAP), a noncanonical form of autophagy, protects against hepatic and systemic inflammation during chronic liver injury in rodents, with beneficial antifibrogenic effects. LAP is enhanced in blood and liver monocytes from patients with fibrosis and cirrhosis. Pharmacological inhibition of LAP components in human monocytes from patients with cirrhosis or genetic disruption of LAP in mice with chronic liver injury exacerbates both the inflammatory signature in isolated human monocytes and the hepatic inflammatory profile in mice, resulting in enhanced liver fibrosis. Mechanistically, patients with cirrhosis showed increased monocyte expression of Fc fragment of IgG receptor IIA (FcγRIIA) and enhanced engulfment of immunoglobulin G in LC3+ phagosomes that triggers an FcγRIIA/Src homology region 2 domain-containing phosphatase-1 (SHP-1) inhibitory immunoreceptor tyrosine-based activation motif (ITAMi) anti-inflammatory pathway. Mice overexpressing human FcγRIIA in myeloid cells show enhanced LAP in response to chronic liver injury and resistance to inflammation and liver fibrosis. Activation of LAP is lost in monocytes from patients with multiorgan failure and restored by specifically targeting ITAMi signaling with anti-FcγRIIA F(ab')2 fragments, or with intravenous immunoglobulin (IVIg). These data suggest the existence of an ITAMi-mediated mechanism by which LAP might protect against inflammation. Sustaining LAP may open therapeutic perspectives for patients with chronic liver disease.
Collapse
Affiliation(s)
- JingHong Wan
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France
| | - Emmanuel Weiss
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France.,Department of Anesthesiology and Critical Care, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, F-92110 Clichy, France
| | - Sanae Ben Mkaddem
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France
| | - Morgane Mabire
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France
| | - Pierre-Marie Choinier
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France.,Department of Anesthesiology and Critical Care, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, F-92110 Clichy, France
| | - Olivia Picq
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France.,Department of Anesthesiology and Critical Care, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, F-92110 Clichy, France
| | - Tristan Thibault-Sogorb
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France.,Department of Anesthesiology and Critical Care, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, F-92110 Clichy, France
| | - Pushpa Hegde
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France
| | - Dorsa Pishvaie
- Department of Hepatology, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, F-92110, Clichy, France
| | - Marcelle Bens
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France
| | - Linda Broer
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France
| | - Hélène Gilgenkrantz
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France
| | - Richard Moreau
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France.,Department of Hepatology, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, F-92110, Clichy, France
| | - Loredana Saveanu
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France
| | - Patrice Codogno
- Université de Paris Institut Necker-Enfants malades (INEM), INSERM, U1151, CNRS UMR8223, F-75015 Paris, France
| | - Renato C Monteiro
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France
| | - Sophie Lotersztajn
- Université de Paris, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, F-75018 Paris, France.
| |
Collapse
|
29
|
Dunne MR, Wagener J, Loeffler J, Doherty DG, Rogers TR. Unconventional T cells - New players in antifungal immunity. Clin Immunol 2021; 227:108734. [PMID: 33895356 DOI: 10.1016/j.clim.2021.108734] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 12/29/2022]
Abstract
Life-threatening invasive fungal diseases (IFD) are increasing in incidence, especially in immunocompromised patients and successful resolution of IFD requires a variety of different immune cells. With the limited repertoire of available antifungal drugs there is a need for more effective therapeutic strategies. This review interrogates the evidence on the human immune response to the main pathogens driving IFD, with a focus on the role of unconventional lymphocytes e.g. natural killer (NK) cells, gamma/delta (γδ) T cells, mucosal associated invariant T (MAIT) cells, invariant natural killer T (iNKT) cells and innate lymphoid cells (ILC). Recent discoveries and new insights into the roles of these novel lymphocyte groups in antifungal immunity will be discussed, and we will explore how an improved understanding of antifungal action by lymphocytes can inform efforts to improve antifungal treatment options.
Collapse
Affiliation(s)
- Margaret R Dunne
- Department of Clinical Microbiology, Trinity College Dublin, Sir Patrick Dun Research Laboratory, St James's Hospital, Dublin 8, Ireland; Department of Immunology, School of Medicine, Trinity College Dublin, Dublin 8, Ireland.
| | - Johannes Wagener
- Department of Clinical Microbiology, Trinity College Dublin, Sir Patrick Dun Research Laboratory, St James's Hospital, Dublin 8, Ireland
| | - Juergen Loeffler
- Department of Internal Medicine II, WÜ4i, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Derek G Doherty
- Department of Immunology, School of Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Thomas R Rogers
- Department of Clinical Microbiology, Trinity College Dublin, Sir Patrick Dun Research Laboratory, St James's Hospital, Dublin 8, Ireland
| |
Collapse
|
30
|
Liu Z, Ding F, Shen X. Total flavonoids of Radix Tetrastigma suppress inflammation-related hepatocellular carcinoma cell metastasis. Mol Genet Genomics 2021; 296:571-579. [PMID: 33576897 PMCID: PMC8144124 DOI: 10.1007/s00438-020-01759-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/21/2020] [Indexed: 11/30/2022]
Abstract
This study aimed to investigate the effects of the total flavonoids of Radix Tetrastigma (RTF) on inflammation-related hepatocellular carcinoma (HCC) development. Extracted RTF was diluted to different concentrations for subsequent experiments. HCC cells were cotreated with lipopolysaccharide (LPS) and RTF to investigate the effects of RTF on LPS-stimulated HCC cells. A CCK-8 kit was used to measure cell proliferation. Apoptosis was detected with a flow cytometer. Cell migration and invasion were quantified by wound healing and Transwell assays, respectively. The expression of TLR4 and COX-2 and activation of the NF-κB pathway were determined by Western blotting. Treatment with LPS significantly enhanced cell proliferation and decreased the apoptosis rate, while cell migration and invasion were notably upregulated. RTF suppressed the proliferation and invasion induced by LPS stimulation and promoted HCC cell apoptosis. The protein levels of Bax and cleaved caspase-3 were decreased and that of Bcl-2 was increased by LPS in HCC cells, which could be rescued by RTF. RTF significantly inhibited the LPS-induced expression of the proinflammatory mediators IL-6 and IL-8 in HCC cells. Mechanistically, with RTF treatment, the upregulated expression of TLR4 and COX-2 induced by LPS was obviously downregulated. Furthermore, the phosphorylation of NF-κB/p65 was significantly decreased in LPS-stimulated cells after supplementation with RTF. Our study suggests that RTF exerts a significant inhibitory effect on the LPS-induced enhancement of the malignant behaviors of HCC cells via inactivation of TLR4/NF-κB signaling. RTF may be a promising chemotherapeutic agent to limit HCC development and inflammation-mediated metastasis.
Collapse
Affiliation(s)
- Zhendong Liu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of TCM), Hangzhou, 310006, China
| | - Fangmi Ding
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of TCM), Hangzhou, 310006, China
| | - Xingyong Shen
- Department of Oncology, Xijing Hospital, Air Force Military Medical University, 15 Changle West Road, Xian, 710032, Shaanxi, China.
| |
Collapse
|
31
|
Kalia N, Singh J, Kaur M. The role of dectin-1 in health and disease. Immunobiology 2021; 226:152071. [PMID: 33588306 DOI: 10.1016/j.imbio.2021.152071] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/07/2021] [Accepted: 01/31/2021] [Indexed: 02/08/2023]
Abstract
Dendritic cell-associated C-type lectin-1 (Dectin-1), also known as β-glucan receptor is an emerging pattern recognition receptor (PRR) which belongs to the family of C-type lectin receptor (CLR). This CLR identifies ligands independently of Ca2+ and is majorly involved in coupling of innate with adaptive immunity. Formerly, Dectin-1 was best known for its role in anti-fungal defense only. However, recent explorations suggested its wider role in defense against variety of infectious diseases caused by pathogens including bacteria, parasites and viruses. In fact, Dectin-1 signaling axis has been suggested to be targeted as an effective therapeutic strategy for cancers. Dectin-1 has also been elucidated ascetically in the heart, respiratory, intestinal, neurological and developmental disorders. Being a defensive PRR, Dectin-1 results in optimal immune responses in collaboration with other PRRs, but the overall evaluation reinforces the hypothesis of disease development on dis-regulation of Dectin-1 activity. This underscores the impact of Dectin-1 polymorphisms in modulating protein expression and generation of non-optimal immune responses through defective collaborations, further underlining their therapeutic potential. To add on, Dectin-1 influence autoimmunity and severe inflammation accredited to recognition of self T cells and apoptotic cells through unknown ligands. Few reports have also testified its redundant role in infections, which makes it a complicated molecule to be fully resolved. Thus, Dectin-1 is a hub that runs a complex collaborative network, whose interactive wire connections to different PRRs are still pending to be revealed. Alternatively, so far focus of almost all the researchers was the two major cell surface isoforms of Dectin-1, despite the fact that its soluble functional intracellular isoform (Dectin-1E) has already been dissected but is indefinable. Therefore, this review intensely recommends the need of future research to resolve the un-resolved and treasure the comprehensive role of Dectin-1 in different clinical outcomes, before determining its therapeutic prospective.
Collapse
Affiliation(s)
- Namarta Kalia
- Department of Molecular Biology & Biochemistry, Guru Nanak Dev University, Amritsar 143001, India.
| | - Jatinder Singh
- Department of Molecular Biology & Biochemistry, Guru Nanak Dev University, Amritsar 143001, India
| | - Manpreet Kaur
- Department of Human Genetics, Guru Nanak Dev University, Amritsar 143001, India.
| |
Collapse
|
32
|
Candida Administration Worsens Uremia-Induced Gut Leakage in Bilateral Nephrectomy Mice, an Impact of Gut Fungi and Organismal Molecules in Uremia. mSystems 2021; 6:6/1/e01187-20. [PMID: 33436518 PMCID: PMC7901485 DOI: 10.1128/msystems.01187-20] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The impact of gut fungi and (1→3)-β-d-glucan (BG), a major fungal cell wall component, on uremia was explored by Candida albicans oral administration in bilateral nephrectomy (BiNx) mice because of the prominence of C. albicans in the human intestine but not in mice. As such, BiNx with Candida administration (BiNx-Candida) enhanced intestinal injury (colon cytokines and apoptosis), gut leakage (fluorescein isothiocyanate [FITC]-dextran assay, endotoxemia, serum BG, and bacteremia), systemic inflammation, and liver injury at 48 h postsurgery compared with non-Candida BiNx mice. Interestingly, uremia-induced enterocyte apoptosis was severe enough for gut translocation of viable bacteria, as indicated by culture positivity for bacteria in blood, mesenteric lymph nodes (MLNs), and other organs, which was more severe in BiNx-Candida than in non-Candida BiNx mice. Candida induced alterations in the gut microbiota of BiNx mice as indicated by (i) the higher fungal burdens in the feces of BiNx-Candida mice than in sham-Candida mice by culture methods and (ii) increased Bacteroides with decreased Firmicutes and reduced bacterial diversity in the feces of BiNx-Candida mice compared with non-Candida BiNx mice by fecal microbiome analysis. In addition, lipopolysaccharide plus BG (LPS+BG), compared with each molecule alone, induced high supernatant cytokine levels, which were enhanced by uremic mouse serum in both hepatocytes (HepG2 cells) and macrophages (RAW264.7 cells). Moreover, LPS+BG, but not each molecule alone, reduced the glycolysis capacity and mitochondrial function in HepG2 cells as determined by extracellular flux analysis. Additionally, a probiotic, Lactobacillus rhamnosus L34 (L34), attenuated disease severity only in BiNx-Candida mice but not in non-Candida BiNx mice, as indicated by liver injury and serum cytokines through the attenuation of gut leakage, the fecal abundance of fungi, and fecal bacterial diversity but not fecal Gram-negative bacteria. In conclusion, Candida enhanced BiNx severity through the worsening of gut leakage and microbiota alterations that resulted in bacteremia, endotoxemia, and glucanemia.IMPORTANCE The impact of fungi in the intestine on acute uremia was demonstrated by the oral administration of Candida albicans in mice with the removal of both kidneys. Because fungi in the mouse intestine are less abundant than in humans, a Candida-administered mouse model has more resemblance to patient conditions. Accordingly, acute uremia, without Candida, induced intestinal mucosal injury, which resulted in the translocation of endotoxin, a major molecule of gut bacteria, from the intestine into blood circulation. In acute uremia with Candida, intestinal injury was more severe due to fungi and the alteration in intestinal bacteria (increased Bacteroides with decreased Firmicutes), leading to the gut translocation of both endotoxin from gut bacteria and (1→3)-β-d-glucan from Candida, which synergistically enhanced systemic inflammation in acute uremia. Both pathogen-associated molecules were delivered to the liver and induced hepatocyte inflammatory responses with a reduced energy production capacity, resulting in acute uremia-induced liver injury. In addition, Lactobacillus rhamnosus attenuated intestinal injury through reduced gut Candida and improved intestinal bacterial conditions.
Collapse
|
33
|
Qi X, Yang M, Stenberg J, Dey R, Fogwe L, Alam MS, Kimchi ET, Staveley-O'Carroll KF, Li G. Gut microbiota mediated molecular events and therapy in liver diseases. World J Gastroenterol 2020; 26:7603-7618. [PMID: 33505139 PMCID: PMC7789060 DOI: 10.3748/wjg.v26.i48.7603] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/24/2020] [Accepted: 12/06/2020] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota is a community of microorganisms that reside in the gastrointestinal tract. An increasing number of studies has demonstrated that the gut-liver axis plays a critical role in liver homeostasis. Dysbiosis of gut microbiota can cause liver diseases, including nonalcoholic fatty liver disease and alcoholic liver disease. Preclinical and clinical investigations have substantiated that the metabolites and other molecules derived from gut microbiota and diet interaction function as mediators to cause liver fibrosis, cirrhosis, and final cancer. This effect has been demonstrated to be associated with dysregulation of intrahepatic immunity and liver metabolism. Targeting these findings have led to the development of novel preventive and therapeutic strategies. Here, we review the cellular and molecular mechanisms underlying gut microbiota-mediated impact on liver disease. We also summarize the advancement of gut microbiota-based therapeutic strategies in the control of liver diseases.
Collapse
Affiliation(s)
- Xiaoqiang Qi
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, United States
- VA Hospital, Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, United States
- VA Hospital, Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States
| | - Joseph Stenberg
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
| | - Rahul Dey
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
| | - Leslie Fogwe
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
| | | | - Eric T Kimchi
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, United States
- VA Hospital, Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States
| | - Kevin F Staveley-O'Carroll
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, United States
- VA Hospital, Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States
| | - Guangfu Li
- Department of Surgery, University of Missouri, Columbia, MO 65212, United States
- Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, United States
- VA Hospital, Harry S Truman Memorial VA Hospital, Columbia, MO 65201, United States
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65212, United States
| |
Collapse
|
34
|
Ouaguia L, Dufeu-Duchesne T, Leroy V, Decaens T, Reiser JB, Sosa Cuevas E, Durantel D, Valladeau-Guilemond J, Bendriss-Vermare N, Chaperot L, Aspord C. Hepatitis B virus exploits C-type lectin receptors to hijack cDC1s, cDC2s and pDCs. Clin Transl Immunology 2020; 9:e1208. [PMID: 33312564 PMCID: PMC7723857 DOI: 10.1002/cti2.1208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 01/07/2023] Open
Abstract
Objectives C‐type lectin receptors (CLRs) are key receptors used by DCs to orchestrate responses to pathogens. During infections, the glycan–lectin interactions shape the virus–host interplay and viruses can subvert the function of CLRs to escape antiviral immunity. Recognition of virus/viral components and uptake by CLRs together with subsequent signalling cascades are crucial in initiating and shaping antiviral immunity, and decisive in the outcome of infection. Yet, the interaction of hepatitis B virus (HBV) with CLRs remains largely unknown. As HBV hijacks DC subsets and viral antigens harbour glycan motifs, we hypothesised that HBV may subvert DCs through CLR binding. Methods We investigated here the pattern of CLR expression on BDCA1+ cDC2s, BDCA2+ pDCs and BDCA3+ cDC1s from both blood and liver of HBV‐infected patients and explored the ability of HBsAg to bind DC subsets through specific CLRs. Results We highlighted for the first time that the CLR repertoire of circulating and intrahepatic cDC2s, cDC1s and pDCs was perturbed in patients with chronic HBV infection and that some CLR expression levels correlated with plasma HBsAg and HBV DNA levels. We also identified candidate CLR responsible for HBsAg binding to cDCs (CD367/DCIR/CLEC4A, CD32/FcɣRIIA) and pDCs (CD369/DECTIN1/CLEC7A, CD336/NKp44) and demonstrated that HBsAg inhibited DC functions in a CLR‐ and glycosylation‐dependent manner. Conclusion HBV may exploit CLR pathways to hijack DC subsets and escape from immune control. Such advances bring insights into the mechanisms by which HBV subverts immunity and pave the way for developing innovative therapeutic strategies to restore an efficient immune control of the infection by manipulating the viral glycan–lectin axis.
Collapse
Affiliation(s)
- Laurissa Ouaguia
- Institute for Advanced Biosciences, Immunobiology and Immunotherapy in Chronic Diseases Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes Grenoble France.,R&D Laboratory Etablissement Français du Sang Auvergne-Rhône-Alpes Grenoble France
| | - Tania Dufeu-Duchesne
- Hepato-Gastroenterology Unit CHU Grenoble Alpes Grenoble France.,Institute for Advanced Biosciences Research Center Inserm U1209/CNRS 5309/UGA Analytic Immunology of Chronic Pathologies La Tronche France
| | - Vincent Leroy
- Hepato-Gastroenterology Unit CHU Grenoble Alpes Grenoble France.,Institute for Advanced Biosciences Research Center Inserm U1209/CNRS 5309/UGA Analytic Immunology of Chronic Pathologies La Tronche France.,Université Grenoble Alpes Grenoble France
| | - Thomas Decaens
- Hepato-Gastroenterology Unit CHU Grenoble Alpes Grenoble France.,Institute for Advanced Biosciences Research Center Inserm U1209/CNRS 5309/UGA Analytic Immunology of Chronic Pathologies La Tronche France.,Université Grenoble Alpes Grenoble France
| | - Jean-Baptiste Reiser
- Institut de Biologie Structurale CNRS CEA Université Grenoble Alpes Grenoble France
| | - Eleonora Sosa Cuevas
- Institute for Advanced Biosciences, Immunobiology and Immunotherapy in Chronic Diseases Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes Grenoble France.,R&D Laboratory Etablissement Français du Sang Auvergne-Rhône-Alpes Grenoble France
| | - David Durantel
- INSERM 1052 CNRS 5286 Centre Léon Bérard Centre de Recherche en Cancérologie de Lyon Université Lyon Université Claude Bernard Lyon 1 Lyon France
| | - Jenny Valladeau-Guilemond
- INSERM 1052 CNRS 5286 Centre Léon Bérard Centre de Recherche en Cancérologie de Lyon Université Lyon Université Claude Bernard Lyon 1 Lyon France
| | - Nathalie Bendriss-Vermare
- INSERM 1052 CNRS 5286 Centre Léon Bérard Centre de Recherche en Cancérologie de Lyon Université Lyon Université Claude Bernard Lyon 1 Lyon France
| | - Laurence Chaperot
- Institute for Advanced Biosciences, Immunobiology and Immunotherapy in Chronic Diseases Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes Grenoble France.,R&D Laboratory Etablissement Français du Sang Auvergne-Rhône-Alpes Grenoble France
| | - Caroline Aspord
- Institute for Advanced Biosciences, Immunobiology and Immunotherapy in Chronic Diseases Inserm U 1209 CNRS UMR 5309 Université Grenoble Alpes Grenoble France.,R&D Laboratory Etablissement Français du Sang Auvergne-Rhône-Alpes Grenoble France
| |
Collapse
|
35
|
Effendi WI, Nagano T, Hasan H, Yudhawati R. Immunoregulatory Property of C-Type Lectin-Like Receptors in Fibrosing Interstitial Lung Diseases. Int J Mol Sci 2020; 21:E3665. [PMID: 32455964 PMCID: PMC7279300 DOI: 10.3390/ijms21103665] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 11/16/2022] Open
Abstract
The innate immune system identifies exogenous threats or endogenous stress through germline-encoded receptors called pattern recognition receptors (PRRs) that initiate consecutive downstream signaling pathways to control immune responses. However, the contribution of the immune system and inflammation to fibrosing interstitial lung diseases (ILD) remains poorly understood. Immunoreceptor tyrosine-based motif-bearing C-type lectin-like receptors (CTLRs) may interact with various immune cells during tissue injury and wound repair processes. Dectin-1 is a CTLR with dominant mechanisms manifested through its intracellular signaling cascades, which regulate fibrosis-promoting properties through gene transcription and cytokine activation. Additionally, immune impairment in ILD facilitates microbiome colonization; hence, Dectin-1 is the master protector in host pulmonary defense against fungal invasion. Recent progress in determining the signaling pathways that control the balance of fibrosis has implicated immunoreceptor tyrosine-based motif-bearing CTLRs as being involved, either directly or indirectly, in the pathogenesis of fibrosing ILD.
Collapse
Affiliation(s)
- Wiwin Is Effendi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan;
- Department of Pulmonology and Respiratory Medicine, Medical Faculty of Airlangga University, Surabaya 60131, Indonesia; (H.H.); (R.Y.)
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan;
| | - Helmia Hasan
- Department of Pulmonology and Respiratory Medicine, Medical Faculty of Airlangga University, Surabaya 60131, Indonesia; (H.H.); (R.Y.)
| | - Resti Yudhawati
- Department of Pulmonology and Respiratory Medicine, Medical Faculty of Airlangga University, Surabaya 60131, Indonesia; (H.H.); (R.Y.)
| |
Collapse
|
36
|
Dermatopontin, A Novel Adipokine Promoting Adipose Tissue Extracellular Matrix Remodelling and Inflammation in Obesity. J Clin Med 2020; 9:jcm9041069. [PMID: 32283761 PMCID: PMC7230369 DOI: 10.3390/jcm9041069] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/27/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022] Open
Abstract
Compelling evidence suggests that dermatopontin (DPT) regulates collagen and fibronectin fibril formation, the induction of cell adhesion and the prompting of wound healing. We aimed to evaluate the role of DPT on obesity and its associated metabolic alterations as well as its impact in visceral adipose tissue (VAT) inflammation and extracellular matrix (ECM) remodelling. Samples obtained from 54 subjects were used in a case-control study. Circulating and VAT expression levels of DPT as well as key ECM remodelling- and inflammation-related genes were analysed. The effect of pro- and anti-inflammatory mediators on the transcript levels of DPT in visceral adipocytes was explored. The impact of DPT on ECM remodelling and inflammation pathways was also evaluated in cultured adipocytes. We show that obesity and obesity-associated type 2 diabetes (T2D) increased (p < 0.05) circulating levels of DPT. In this line, DPT mRNA in VAT was increased (p < 0.05) in obese patients with and without T2D. Gene expression levels of DPT were enhanced (p < 0.05) in human visceral adipocytes after the treatment with lipopolysaccharide, tumour growth factor (TGF)-β and palmitic acid, whereas a downregulation (p < 0.05) was detected after the stimulation with interleukin (IL)-4 and IL-13, critical cytokines mediating anti-inflammatory pathways. Additionally, we revealed that DPT increased (p < 0.05) the expression of ECM- (COL6A3, ELN, MMP9, TNMD) and inflammation-related factors (IL6, IL8, TNF) in human visceral adipocytes. These findings provide, for the first time, evidence of a novel role of DPT in obesity and its associated comorbidities by influencing AT remodelling and inflammation.
Collapse
|
37
|
Hepatocarcinoma-intestine-pancreas/pancreatitis-associated protein (HIP/PAP) confers protection against hepatic fibrosis through downregulation of transforming growth factor β receptor II. J Transl Med 2020; 100:466-482. [PMID: 31641222 DOI: 10.1038/s41374-019-0314-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/23/2019] [Accepted: 07/30/2019] [Indexed: 01/18/2023] Open
Abstract
Hepatocarcinoma-intestine-pancreas/pancreatitis-associated protein (HIP/PAP) has antimicrobial, antioxidant, anti-inflammatory, mitogenic, and antiapoptotic effects and thus exerts important functions in the maintenance of integrity and homeostasis of several organs, such as the gastrointestinal tract, pancreas, and liver. Although the potent hepatoprotective effect of HIP/PAP has been validated, its impact on liver fibrosis has not been reported. In this study, we evaluated the role of HIP/PAP on hepatic fibrosis and explored the possible underlying mechanisms. We found that the expression of HIP/PAP and its mouse counterpart, Reg3B, was markedly upregulated in fibrotic human or mouse livers. Intraperitoneal (i.p.) interleukin (IL)-10, IL-6, and TNF-α but not TGF-β1 significantly induced hepatic overexpression of Reg3B in mice. In both CCl4 and BDL liver fibrosis models, adenovirus-mediated ectopic expression of HIP/PAP markedly alleviated liver injury, inflammation, collagen deposition, hepatic stellate cell activation, and the overexpression of profibrotic cytokines, including transforming growth factor β1 (TGF-β1), platelet-derived growth factor (PDGF)-A, B, connective tissue growth factor (CTGF), and plasminogen activator inhibitor-1 (PAI-1), in mice. In vitro experiments demonstrated that, in addition to suppressing hepatic stellate cell proliferation and accelerating hepatocyte proliferation, HIP/PAP mitigated TGF-β1-induced hepatic stellate cell activation, hepatocyte epithelial-mesenchymal transition (EMT) and upregulated expression of profibrotic cytokines in both hepatic stellate cells and hepatocytes. Moreover, HIP/PAP attenuated the overexpression of TGF-β receptor II (TGF-βRII) in fibrotic mouse livers and decreased the basal expression of TGF-βRII in nonfibrotic mouse livers as well as in cultured hepatocytes and hepatic stellate cells, which is at least partly attributable to the TGF-β1-antagonizing function of HIP/PAP. This study indicates that increased expression of hepatic HIP/PAP serves as a countermeasure against liver injury and fibrosis. Exogenous supplementation of HIP/PAP might be a promising therapeutic agent for hepatic fibrosis as well as liver injury.
Collapse
|
38
|
Li X, Bian Y, Pang P, Yu S, Wang X, Gao Y, Liu K, Liu Q, Yuan Y, Du W. Inhibition of Dectin-1 in mice ameliorates cardiac remodeling by suppressing NF-κB/NLRP3 signaling after myocardial infarction. Int Immunopharmacol 2020; 80:106116. [PMID: 31978804 DOI: 10.1016/j.intimp.2019.106116] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/16/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022]
Abstract
The myocardial inflammatory response is a consequence of myocardial infarction (MI), which may deteriorate cardiac remodeling and lead to dysfunction in the heart post-MI. Dectin-1 is a c-type lectin, which has been shown to regulate innate immune responses to pathogens. However, the role of Dectin-1 in the heart diseases remains largely unknown. In this study, we aimed to investigate the effects of Dectin-1 on cardiac remodeling post-MI. We found that cardiac Dectin-1 mRNA and protein expressions were significantly elevated in C57BL/6 mice after MI. In vitro, hypoxia induced cardiomyocyte injury in parallel with increased Dectin-1 protein expression. Knockdown of Dectin-1 remarkably attenuated cardiomyocyte death under hypoxia and lipopolysaccharide (LPS) stimulation. In vivo administration of adeno-associated virus serotype 9 mediated silencing of Dectin-1, which significantly decreased cardiac fibrosis, dilatation, and improved cardiac function in the mice post-MI. At the molecular level, downregulation of Dectin-1 dramatically suppressed up-regulation of nuclear factor-κB (NF-κB), nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3), and the inflammatory genes involved in fibrogenesis and cardiac remodeling after MI. Furthermore, treatment with BAY11-7082, an inhibitor of NF-κB, repressed the activation of NF-κB, and attenuated LPS induced elevation of NLRP3 and cell death in cardiomyocytes. Collectively, upregulation of Dectin-1 in cardiomyocytes post-MI contributes to cardiac remodeling and cardiac dysfunction at least partially by activating NF-κB and NLRP3. This study identified Dectin-1 as a promising therapeutic target for ischemic heart disease.
Collapse
Affiliation(s)
- Xin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Yu Bian
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Ping Pang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Shuting Yu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Xiuzhu Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Yuelin Gao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Kuiwu Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Qian Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China
| | - Ye Yuan
- Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150086, China.
| | - Weijie Du
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, PR China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin 150081, PR China.
| |
Collapse
|
39
|
Chen D, Le TH, Shahidipour H, Read SA, Ahlenstiel G. The Role of Gut-Derived Microbial Antigens on Liver Fibrosis Initiation and Progression. Cells 2019; 8:E1324. [PMID: 31717860 PMCID: PMC6912265 DOI: 10.3390/cells8111324] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/12/2022] Open
Abstract
Intestinal dysbiosis has recently become known as an important driver of gastrointestinal and liver disease. It remains poorly understood, however, how gastrointestinal microbes bypass the intestinal mucosa and enter systemic circulation to enact an inflammatory immune response. In the context of chronic liver disease (CLD), insults that drive hepatic inflammation and fibrogenesis (alcohol, fat) can drastically increase intestinal permeability, hence flooding the liver with gut-derived microbiota. Consequently, this may result in exacerbated liver inflammation and fibrosis through activation of liver-resident Kupffer and stellate cells by bacterial, viral, and fungal antigens transported to the liver via the portal vein. This review summarizes the current understanding of microbial translocation in CLD, the cell-specific hepatic response to intestinal antigens, and how this drives the development and progression of hepatic inflammation and fibrosis. Further, we reviewed current and future therapies targeting intestinal permeability and the associated, potentially harmful anti-microbial immune response with respect to their potential in terms of limiting the development and progression of liver fibrosis and end-stage cirrhosis.
Collapse
Affiliation(s)
- Dishen Chen
- Storr Liver Centre, The Westmead Institute for Medical Research, University of Sydney, Westmead 2145, NSW, Australia; (D.C.); (T.H.L.); (H.S.)
| | - Thanh H. Le
- Storr Liver Centre, The Westmead Institute for Medical Research, University of Sydney, Westmead 2145, NSW, Australia; (D.C.); (T.H.L.); (H.S.)
- School of Medicine, Western Sydney University, Campbelltown 2560, NSW, Australia
| | - Haleh Shahidipour
- Storr Liver Centre, The Westmead Institute for Medical Research, University of Sydney, Westmead 2145, NSW, Australia; (D.C.); (T.H.L.); (H.S.)
- Blacktown Medical School, Western Sydney University, Blacktown 2148, NSW, Australia
| | - Scott A. Read
- Storr Liver Centre, The Westmead Institute for Medical Research, University of Sydney, Westmead 2145, NSW, Australia; (D.C.); (T.H.L.); (H.S.)
- Blacktown Medical School, Western Sydney University, Blacktown 2148, NSW, Australia
| | - Golo Ahlenstiel
- Storr Liver Centre, The Westmead Institute for Medical Research, University of Sydney, Westmead 2145, NSW, Australia; (D.C.); (T.H.L.); (H.S.)
- Blacktown Medical School, Western Sydney University, Blacktown 2148, NSW, Australia
- Blacktown Hospital, Blacktown 2148, NSW, Australia
| |
Collapse
|
40
|
Paramylon, a Potent Immunomodulator from WZSL Mutant of Euglena gracilis. Molecules 2019; 24:molecules24173114. [PMID: 31461965 PMCID: PMC6749516 DOI: 10.3390/molecules24173114] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/08/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023] Open
Abstract
β-glucans, heterogeneous glucose polymers present in many organisms, have the capability to activate the innate immune system. Efficacy of activation depends on purity of the compound, molecular structure, polymerization degree, and source. One of the purest forms of crystallized β-(1–3)-glucan present in nature is the paramylon extracted from the WZSL non-chloroplastic mutant of Euglena gracilis, which can be processed to produce linear nanofibers capable of interacting with specific receptors present on cell membranes. The effects of these nanofibers, already investigated on plants, animals, and humans, will be analyzed.
Collapse
|
41
|
Abstract
The C-type lectins are a superfamily of proteins that recognize a broad repertoire of ligands and that regulate a diverse range of physiological functions. Most research attention has focused on the ability of C-type lectins to function in innate and adaptive antimicrobial immune responses, but these proteins are increasingly being recognized to have a major role in autoimmune diseases and to contribute to many other aspects of multicellular existence. Defects in these molecules lead to developmental and physiological abnormalities, as well as altered susceptibility to infectious and non-infectious diseases. In this Review, we present an overview of the roles of C-type lectins in immunity and homeostasis, with an emphasis on the most exciting recent discoveries.
Collapse
|
42
|
Li TH, Liu L, Hou YY, Shen SN, Wang TT. C-type lectin receptor-mediated immune recognition and response of the microbiota in the gut. Gastroenterol Rep (Oxf) 2019; 7:312-321. [PMID: 31687150 PMCID: PMC6821170 DOI: 10.1093/gastro/goz028] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 06/05/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023] Open
Abstract
C-type lectin receptors (CLRs) are powerful pattern-recognition receptors that discern ‘self’ and ‘non-self’ in our body and protect us from invasive pathogens by mediating immune recognition and response. The gastrointestinal tract is very important for the maintenance of homeostasis; it is the largest shelter for the billions of microorganisms in the body and CLRs play a crucial regulatory role in this system. This study focuses on several CLRs, including Dectin-1, Dectin-2, Dectin-3 and Mincle. We summarize the roles of CLRs in maintaining gastrointestinal immune-system homeostasis, especially their functions in mediating immune recognition and responses in the gut, discuss their relationships to some diseases, highlight the significance of CLR-mediated sensing of microbial and non-microbial compounds in the gut immune system and identify new therapeutic targets.
Collapse
Affiliation(s)
- Tian-Hang Li
- Immunology and Reproduction Biology Lab, Medical School of Nanjing University, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, P. R. China
| | - Ling Liu
- Immunology and Reproduction Biology Lab, Medical School of Nanjing University, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, P. R. China
| | - Ya-Yi Hou
- Immunology and Reproduction Biology Lab, Medical School of Nanjing University, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, P. R. China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, P. R. China
| | - Su-Nan Shen
- Immunology and Reproduction Biology Lab, Medical School of Nanjing University, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, P. R. China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, P. R. China
| | - Ting-Ting Wang
- Immunology and Reproduction Biology Lab, Medical School of Nanjing University, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, P. R. China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, P. R. China
| |
Collapse
|
43
|
Tang C, Makusheva Y, Sun H, Han W, Iwakura Y. Myeloid C-type lectin receptors in skin/mucoepithelial diseases and tumors. J Leukoc Biol 2019; 106:903-917. [PMID: 30964564 PMCID: PMC6850291 DOI: 10.1002/jlb.2ri0119-031r] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/11/2019] [Accepted: 03/20/2019] [Indexed: 12/30/2022] Open
Abstract
Myeloid C‐type lectin receptors (CLRs), which consist of an extracellular carbohydrate recognition domain and intracellular signal transducing motif such as the immunoreceptor tyrosine‐based activation motif (ITAM) or immunoreceptor tyrosine‐based inhibitory motif (ITIM), are innate immune receptors primarily expressed on myeloid lineage cells such as dendritic cells (DCs) and Mϕs. CLRs play important roles in host defense against infection by fungi and bacteria by recognizing specific carbohydrate components of these pathogens. However, these immune receptors also make important contributions to immune homeostasis of mucosa and skin in mammals by recognizing components of microbiota, as well as by recognizing self‐components such as alarmins from dead cells and noncanonical non‐carbohydrate ligands. CLR deficiency not only induces hypersensitivity to infection, but also causes dysregulation of muco‐cutaneous immune homeostasis, resulting in the development of allergy, inflammation, autoimmunity, and tumors. In this review, we introduce recent discoveries regarding the roles of myeloid CLRs in the immune system exposed to the environment, and discuss the roles of these lectin receptors in the development of colitis, asthma, psoriasis, atopic dermatitis, and cancer. Although some CLRs are suggested to be involved in the development of these diseases, the function of CLRs and their ligands still largely remain to be elucidated.
Collapse
Affiliation(s)
- Ce Tang
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yulia Makusheva
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Haiyang Sun
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Wei Han
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| |
Collapse
|
44
|
Schemitt EG, Hartmann RM, Colares JR, Licks F, Salvi JO, Marroni CA, Marroni NP. Protective action of glutamine in rats with severe acute liver failure. World J Hepatol 2019; 11:273-286. [PMID: 30967905 PMCID: PMC6447424 DOI: 10.4254/wjh.v11.i3.273] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/29/2019] [Accepted: 03/12/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Severe acute liver failure (SALF) is a rare, but high-mortality, rapidly evolving syndrome that leads to hepatocyte degeneration with impaired liver function. Thioacetamide (TAA) is a known xenobiotic, which promotes the increase of the formation of reactive oxygen species. Erythroid 2-related factor 2 (Nrf2) activates the antioxidant protection of cells. Studies have evidenced the involvement of inflammatory mediators in conditions of oxidative stress. AIM To evaluate the antioxidant effects of glutamine on Nrf2 activation and NFκB-mediated inflammation in rats with TAA-induced IHAG. METHODS Male Wistar rats (n = 28) were divided into four groups: control, control+glutamine, TAA, and TAA + glutamine. Two TAA doses (400 mg/kg) were administered intraperitoneally, 8 h apart. Glutamine (25 mg/kg) was administered at 30 min, 24 h, and 36 h. At 48 h, blood was collected for liver integrity analysis [aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP)]. The liver was harvested for histology and assessment of oxidative stress [thiobarbituric acid-reactive substances (TBARS), catalase (CAT), glutathione peroxidase (GPx), glutathione S-transferase (GST), glutathione (GSH), Nrf2, Kelch-like ECH-associated protein 1 (Keap1), NADPH quinone oxidoreductase1 (NQO1), superoxide dismutase (SOD)] and inflammatory process. RESULTS TAA caused disruption of the hepatic parenchyma, with inflammatory infiltration, massive necrosis, and ballooning degeneration. Glutamine mitigated this tissue damage, with visible regeneration of hepatic parenchyma; decreased TBARS (P < 0.001), GSH (P < 0.01), IL-1β, IL6, and TNFα levels (P <0.01) in hepatic tissue; and decreased blood levels of AST, ALT, and ALP (P <0.05). In addition, CAT, GPx, and GST activities were restored in the glutamine group (P <0.01, P <0.01, and P <0.001, respectively vs TAA alone). Glutamine increased expression of Nrf2 (P < 0.05), NQO1, and SOD (P < 0.01), as well as levels of IL-10 (P <0.001), while decreasing expression of Keap1, TLR4, NFκB (P < 0.001), COX-2 and iNOS, (P < 0.01), and reducing NO2 and NO3 levels (P < 0.05). CONCLUSION In the TAA experimental model of IHAG, glutamine activated the Nrf2 pathway, thus promoting antioxidant protection, and blunted the NFκB-mediated pathway, reducing inflammation.
Collapse
Affiliation(s)
- Elizângela G Schemitt
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| | - Renata M Hartmann
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| | - Josieli R Colares
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| | - Francielli Licks
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| | - Jéferson O Salvi
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| | - Cláudio A Marroni
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil.
| | - Norma P Marroni
- Laboratory of Experimental Hepatology and Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre 90040060, Brazil
| |
Collapse
|
45
|
Zhang S, Guo S, Gao XB, Liu A, Jiang W, Chen X, Yang P, Liu LN, Shi L, Zhang Y. Matrine attenuates high-fat diet-induced in vivo and ox-LDL-induced in vitro vascular injury by regulating the PKCα/eNOS and PI3K/Akt/eNOS pathways. J Cell Mol Med 2019; 23:2731-2743. [PMID: 30770623 PMCID: PMC6433715 DOI: 10.1111/jcmm.14180] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 11/15/2018] [Accepted: 01/04/2019] [Indexed: 12/14/2022] Open
Abstract
Lipid metabolism disorders lead to vascular endothelial injury. Matrine is an alkaloid that has been used to improve obesity and diabetes and for the treatment of hepatitis B. However, its effect on lipid metabolism disorders and vascular injury is unclear. Here, we investigated the effect of matrine on high‐fat diet fed mice and oxidized low‐density lipoprotein (ox‐LDL)‐induced human umbilical vein endothelial cells (HUVECs). Computational virtual docking analyses, phosphoinositide 3‐kinase (PI3K) and protein kinase C‐α (PKCα) inhibitors were used to localize matrine in vascular injuries. The results showed that matrine‐treated mice were more resistant to abnormal lipid metabolism and inflammation than vehicle‐treated mice and exhibited significantly alleviated ox‐LDL‐stimulated dysfunction of HUVECs, restored diminished nitric oxide release, decreased reactive oxygen species generation and increased expression phosphorylation of AKT‐Ser473 and endothelial nitric oxide synthase (eNOS)‐Ser1177. Matrine not only up‐regulates eNOS‐Ser1177 but also down‐regulates eNOS‐Thr495, a PKCα‐controlled negative regulator of eNOS. Using computational virtual docking analyses and biochemical assays, matrine was also shown to influence eNOS/NO via PKCα inhibition. Moreover, the protective effects of matrine were significantly abolished by the simultaneous application of PKCα and the PI3K inhibitor. Matrine may thus be potentially employed as a novel therapeutic strategy against high‐fat diet‐induced vascular injury.
Collapse
Affiliation(s)
- Song Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Shun Guo
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Xiao-Bo Gao
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - An Liu
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Wei Jiang
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Xi Chen
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Peng Yang
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Lin-Na Liu
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Lei Shi
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| | - Yan Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, PR China
| |
Collapse
|
46
|
Torres-Hernandez A, Wang W, Nikiforov Y, Tejada K, Torres L, Kalabin A, Wu Y, Haq MIU, Khan MY, Zhao Z, Su W, Camargo J, Hundeyin M, Diskin B, Adam S, Rossi JAK, Kurz E, Aykut B, Shadaloey SAA, Leinwand J, Miller G. Targeting SYK signaling in myeloid cells protects against liver fibrosis and hepatocarcinogenesis. Oncogene 2019; 38:4512-4526. [PMID: 30742098 DOI: 10.1038/s41388-019-0734-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 02/07/2023]
Abstract
Liver fibrosis and fibrosis-associated hepatocarcinogenesis are driven by chronic inflammation and are leading causes of morbidity and death worldwide. SYK signaling regulates critical processes in innate and adaptive immunity, as well as parenchymal cells. We discovered high SYK expression in the parenchymal hepatocyte, hepatic stellate cell (HSC), and the inflammatory compartments in the fibrotic liver. We postulated that targeting SYK would mitigate hepatic fibrosis and oncogenic progression. We found that inhibition of SYK with the selective small molecule inhibitors Piceatannol and PRT062607 markedly protected against toxin-induced hepatic fibrosis, associated hepatocellular injury and intra-hepatic inflammation, and hepatocarcinogenesis. SYK inhibition resulted in increased intra-tumoral expression of the p16 and p53 but decreased expression of Bcl-xL and SMAD4. Further, hepatic expression of genes regulating angiogenesis, apoptosis, cell cycle regulation, and cellular senescence were affected by targeting SYK. We found that SYK inhibition mitigated both HSC trans-differentiation and acquisition of an inflammatory phenotype in T cells, B cells, and myeloid cells. However, in vivo experiments employing selective targeted deletion of SYK indicated that only SYK deletion in the myeloid compartment was sufficient to confer protection against fibrogenic progression. Targeting SYK promoted myeloid cell differentiation into hepato-protective TNFαlow CD206hi phenotype downregulating mTOR, IL-8 signaling and oxidative phosphorylation. Collectively, these data suggest that SYK is an attractive target for experimental therapeutics in treating hepatic fibrosis and oncogenesis.
Collapse
Affiliation(s)
- Alejandro Torres-Hernandez
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Wei Wang
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Yuri Nikiforov
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Karla Tejada
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Luisana Torres
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Aleksandr Kalabin
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Yue Wu
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Muhammad Israr Ul Haq
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Mohammed Y Khan
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Zhen Zhao
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Wenyu Su
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Jimmy Camargo
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Mautin Hundeyin
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Brian Diskin
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Salma Adam
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Juan A Kochen Rossi
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Emma Kurz
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Berk Aykut
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Sorin A A Shadaloey
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - Joshua Leinwand
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA
| | - George Miller
- S.A. Localio Laboratory, Departments of Surgery, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA. .,Departments of Cell Biology, New York University School of Medicine, 450 East 29th Street, New York, NY, 10016, USA.
| |
Collapse
|
47
|
High-mobility group box 1 induces endoplasmic reticulum stress and activates hepatic stellate cells. J Transl Med 2018; 98:1200-1210. [PMID: 29959419 DOI: 10.1038/s41374-018-0085-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 04/23/2018] [Accepted: 04/25/2018] [Indexed: 12/17/2022] Open
Abstract
Liver fibrosis is a worldwide clinical issue. The activation of hepatic stellate cells (HSCs) is the central event during the hepatic fibrotic response. However, the exact mechanisms related to HSC activation and the connection between hepatocytes and HSCs remain unclear. We elucidated the mechanism by which the nuclear-damage-associated molecular pattern molecule, high-mobility group box 1 (HMGB1) was released from the impaired hepatocytes and induced endoplasmic reticulum stress to activate HSCs. In this work, we demonstrated that HMGB1 can be released from hepatocytes and the released HMGB1 activates the HSCs via ER stress at the transcriptional level which was dependent on the activation of both the TLR4 and RAGE signaling pathways rather than the TLR2 signaling pathway. HMGB1 induction of proinflammatory cytokines interleukin (IL)-1β and IL-18 release was dependent on ER stress. In vivo, stable inhibition of HMGB1 suppressed liver fibrosis. These results suggest that under damage condition, HMGB1 can be secreted from injured hepatocytes and activates TLR4- and RAGE signaling pathways to induce ER stress which activates HSCs. Moreover, HMGB1 can produce multiple inflammatory mediators through ER stress, which, in turn, promote liver fibrosis.
Collapse
|
48
|
Genetic ablation of pannexin1 counteracts liver fibrosis in a chemical, but not in a surgical mouse model. Arch Toxicol 2018; 92:2607-2627. [PMID: 29987408 DOI: 10.1007/s00204-018-2255-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 07/03/2018] [Indexed: 02/07/2023]
Abstract
Liver fibrosis is the final common pathway for almost all causes of chronic liver injury. This chronic disease is characterized by excessive deposition of extracellular matrix components mainly due to transdifferentiation of quiescent hepatic stellate cell into myofibroblasts-like cells, which in turn is driven by cell death and inflammation. In the last few years, paracrine signaling through pannexin1 channels has emerged as a key player in the latter processes. The current study was set up to investigate the role of pannexin1 signaling in liver fibrosis. Wild-type and whole body pannexin1 knock-out mice were treated with carbon tetrachloride or subjected to bile duct ligation. Evaluation of the effects of pannexin1 deletion was based on a number of clinically relevant read-outs, including markers of liver damage, histopathological analysis, oxidative stress, inflammation and regenerative capacity. In parallel, to elucidate the molecular pathways affected by pannexin1 deletion as well as to mechanistically anchor the clinical observations, whole transcriptome analysis of liver tissue was performed. While pannexin1 knock-out mice treated with carbon tetrachloride displayed reduced collagen content, hepatic stellate cell activation, inflammation and hepatic regeneration, bile duct ligated counterparts showed increased hepatocellular injury and antioxidant enzyme activity with a predominant immune response. Gene expression profiling revealed a downregulation of fibrotic and immune responses in pannexin1 knock-out mice treated with carbon tetrachloride, whereas bile duct ligated pannexin1-deficient animals showed a pronounced inflammatory profile. This study shows for the first time an etiology-dependent role for pannexin1 signaling in experimental liver fibrosis.
Collapse
|
49
|
Anti-fibrotic effect of paramylon nanofibers from the WZSL mutant of Euglena gracilis on liver damage induced by CCl 4 in mice. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.05.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
50
|
Del Fresno C, Iborra S, Saz-Leal P, Martínez-López M, Sancho D. Flexible Signaling of Myeloid C-Type Lectin Receptors in Immunity and Inflammation. Front Immunol 2018; 9:804. [PMID: 29755458 PMCID: PMC5932189 DOI: 10.3389/fimmu.2018.00804] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/03/2018] [Indexed: 12/19/2022] Open
Abstract
Myeloid C-type lectin receptors (CLRs) are important sensors of self and non-self that work in concert with other pattern recognition receptors (PRRs). CLRs have been previously classified based on their signaling motifs as activating or inhibitory receptors. However, specific features of the ligand binding process may result in distinct signaling through a single motif, resulting in the triggering of non-canonical pathways. In addition, CLR ligands are frequently exposed in complex structures that simultaneously bind different CLRs and other PRRs, which lead to integration of heterologous signaling among diverse receptors. Herein, we will review how sensing by myeloid CLRs and crosstalk with heterologous receptors is modulated by many factors affecting their signaling and resulting in differential outcomes for immunity and inflammation. Finding common features among those flexible responses initiated by diverse CLR-ligand partners will help to harness CLR function in immunity and inflammation.
Collapse
Affiliation(s)
- Carlos Del Fresno
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Salvador Iborra
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Department of Immunology, School of Medicine, Universidad Complutense de Madrid, 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Paula Saz-Leal
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - María Martínez-López
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| |
Collapse
|