1
|
Jeon D, Hill E, McNeel DG. Toll-like receptor agonists as cancer vaccine adjuvants. Hum Vaccin Immunother 2024; 20:2297453. [PMID: 38155525 PMCID: PMC10760790 DOI: 10.1080/21645515.2023.2297453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy to treat cancer patients. Among the wide range of immunological approaches, cancer vaccines have been investigated to activate and expand tumor-reactive T cells. However, most cancer vaccines have not shown significant clinical benefit as monotherapies. This is likely due to the antigen targets of vaccines, "self" proteins to which there is tolerance, as well as to the immunosuppressive tumor microenvironment. To help circumvent immune tolerance and generate effective immune responses, adjuvants for cancer vaccines are necessary. One representative adjuvant family is Toll-Like receptor (TLR) agonists, synthetic molecules that stimulate TLRs. TLRs are the largest family of pattern recognition receptors (PRRs) that serve as the sensors of pathogens or cellular damage. They recognize conserved foreign molecules from pathogens or internal molecules from cellular damage and propel innate immune responses. When used with vaccines, activation of TLRs signals an innate damage response that can facilitate the development of a strong adaptive immune response against the target antigen. The ability of TLR agonists to modulate innate immune responses has positioned them to serve as adjuvants for vaccines targeting infectious diseases and cancers. This review provides a summary of various TLRs, including their expression patterns, their functions in the immune system, as well as their ligands and synthetic molecules developed as TLR agonists. In addition, it presents a comprehensive overview of recent strategies employing different TLR agonists as adjuvants in cancer vaccine development, both in pre-clinical models and ongoing clinical trials.
Collapse
Affiliation(s)
- Donghwan Jeon
- Department of Oncology, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Ethan Hill
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Douglas G. McNeel
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
2
|
Marwedel B, Medina LY, De May H, Adogla JE, Kennedy E, Flores E, Lim E, Adams S, Bartee E, Serda RE. Regional immune mechanisms enhance efficacy of an autologous cellular cancer vaccine with intraperitoneal administration. Oncoimmunology 2024; 13:2421029. [PMID: 39625271 PMCID: PMC11540083 DOI: 10.1080/2162402x.2024.2421029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 12/06/2024] Open
Abstract
Widespread peritoneal dissemination is common in patients with gynecologic or gastrointestinal cancers. Accumulating evidence of a central role for regional immunity in cancer control indicates that intraperitoneal immunotherapy may have treatment advantages. This study delineates immune mechanisms engaged by intraperitoneal delivery of a cell-based vaccine comprised of silicified ovarian cancer cells associated with enhanced survival. Vaccine trafficking from the site of injection to milky spots and other fat-associated lymphoid clusters was studied in syngeneic cancer models using bioluminescent and fluorescent imaging, microscopy, and flow cytometry. Spectral flow cytometry was used to phenotype peritoneal immune cell populations, while bioluminescent imaging of cancer was used to study myeloid and T cell dependency, systemic immunity, and vaccine efficacy in models of disseminated high-grade serous ovarian and DNA mismatch-repair proficient microsatellite-stable colorectal cancer. Following intraperitoneal vaccination of mice with ovarian cancer, vaccine cells were rapidly internalized by myeloid cells, with subsequent trafficking to fat-associated lymphoid clusters. Tumor clearance was confirmed to be T cell-mediated, leading to the establishment of local and systemic immunity. Combination immune checkpoint inhibitor and vaccine therapy in mice with advanced disease, characterized by an established suppressive tumor microenvironment, increased the number of mice with non-detectable tumors, however, change in tumor burden compared to vaccine monotherapy was not significant. Vaccination also resulted in tumor clearance in mouse models of metastatic colorectal cancer. This study demonstrates that intraperitoneal vaccine delivery has the potential to enhance vaccine efficacy by activating resident immune cells with the subsequent establishment of protective systemic anti-tumor immunity.
Collapse
Affiliation(s)
- Ben Marwedel
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Lorél Y. Medina
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Henning De May
- Department of Obstetrics & Gynecology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Joshua E. Adogla
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Ellie Kennedy
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Erica Flores
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Eunju Lim
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Sarah Adams
- Department of Obstetrics & Gynecology, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Eric Bartee
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| | - Rita E. Serda
- Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM, USA
| |
Collapse
|
3
|
Alhamdan F, Bayarsaikhan G, Yuki K. Toll-like receptors and integrins crosstalk. Front Immunol 2024; 15:1403764. [PMID: 38915411 PMCID: PMC11194410 DOI: 10.3389/fimmu.2024.1403764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/24/2024] [Indexed: 06/26/2024] Open
Abstract
Immune system recognizes invading microbes at both pathogen and antigen levels. Toll-like receptors (TLRs) play a key role in the first-line defense against pathogens. Major functions of TLRs include cytokine and chemokine production. TLRs share common downstream signaling pathways with other receptors. The crosstalk revolving around TLRs is rather significant and complex, underscoring the intricate nature of immune system. The profiles of produced cytokines and chemokines via TLRs can be affected by other receptors. Integrins are critical heterodimeric adhesion molecules expressed on many different cells. There are studies describing synergetic or inhibitory interplay between TLRs and integrins. Thus, we reviewed the crosstalk between TLRs and integrins. Understanding the nature of the crosstalk could allow us to modulate TLR functions via integrins.
Collapse
Affiliation(s)
- Fahd Alhamdan
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia, Boston Children’s Hospital, Boston, MA, United States
- Department of Anesthesia and Immunology, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Ganchimeg Bayarsaikhan
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia, Boston Children’s Hospital, Boston, MA, United States
- Department of Anesthesia and Immunology, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia, Boston Children’s Hospital, Boston, MA, United States
- Department of Anesthesia and Immunology, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
4
|
Duan M, Zhang X, Lou Y, Feng J, Guo P, Ye S, Lv P, Chen Y. Deletion of Tmem268 in mice suppresses anti-infectious immune responses by downregulating CD11b signaling. EMBO Rep 2024; 25:2550-2570. [PMID: 38730209 PMCID: PMC11169502 DOI: 10.1038/s44319-024-00141-6] [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: 05/24/2023] [Revised: 02/25/2024] [Accepted: 04/03/2024] [Indexed: 05/12/2024] Open
Abstract
Transmembrane protein 268 (TMEM268) is a novel, tumor growth-related protein first reported by our laboratory. It interacts with the integrin subunit β4 (ITGB4) and plays a positive role in the regulation of the ITGB4/PLEC signaling pathway. Here, we investigated the effects and mechanism of TMEM268 in anti-infectious immune response in mice. Tmem268 knockout in mice aggravated cecal ligation and puncture-induced sepsis, as evidenced by higher bacterial burden in various tissues and organs, congestion, and apoptosis. Moreover, Tmem268 deficiency in mice inhibited phagocyte adhesion and migration, thus decreasing phagocyte infiltration at the site of infection and complement-dependent phagocytosis. Further findings indicated that TMEM268 interacts with CD11b and inhibits its degradation via the endosome-lysosome pathway. Our results reveal a positive regulatory role of TMEM268 in β2 integrin-associated anti-infectious immune responses and signify the potential value of targeting the TMEM268-CD11b signaling axis for the maintenance of immune homeostasis and immunotherapy for sepsis and related immune disorders.
Collapse
Affiliation(s)
- Mengyuan Duan
- Department of Immunology, Peking University School of Basic Medical Sciences; NHC Key Laboratory of Medical Immunology, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Xuan Zhang
- Department of Immunology, Peking University School of Basic Medical Sciences; NHC Key Laboratory of Medical Immunology, Peking University, 38 Xueyuan Road, 100191, Beijing, China
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Beijing Pediatric Research Institute, Capital Medical University, National Center for Children's Health, 100045, Beijing, China
| | - Yaxin Lou
- Medical and Healthy Analytical Center, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Jinqiu Feng
- Department of Immunology, Peking University School of Basic Medical Sciences; NHC Key Laboratory of Medical Immunology, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Pengli Guo
- Department of Immunology, Peking University School of Basic Medical Sciences; NHC Key Laboratory of Medical Immunology, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Shufang Ye
- Department of Immunology, Peking University School of Basic Medical Sciences; NHC Key Laboratory of Medical Immunology, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Ping Lv
- Department of Immunology, Peking University School of Basic Medical Sciences; NHC Key Laboratory of Medical Immunology, Peking University, 38 Xueyuan Road, 100191, Beijing, China
| | - Yingyu Chen
- Department of Immunology, Peking University School of Basic Medical Sciences; NHC Key Laboratory of Medical Immunology, Peking University, 38 Xueyuan Road, 100191, Beijing, China.
- Center for Human Disease Genomics, Peking University, 38 Xueyuan Road, 100191, Beijing, China.
| |
Collapse
|
5
|
Klaus T, Hieber C, Bros M, Grabbe S. Integrins in Health and Disease-Suitable Targets for Treatment? Cells 2024; 13:212. [PMID: 38334604 PMCID: PMC10854705 DOI: 10.3390/cells13030212] [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: 12/27/2023] [Revised: 01/13/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
Integrin receptors are heterodimeric surface receptors that play multiple roles regarding cell-cell communication, signaling, and migration. The four members of the β2 integrin subfamily are composed of an alternative α (CD11a-d) subunit, which determines the specific receptor properties, and a constant β (CD18) subunit. This review aims to present insight into the multiple immunological roles of integrin receptors, with a focus on β2 integrins that are specifically expressed by leukocytes. The pathophysiological role of β2 integrins is confirmed by the drastic phenotype of patients suffering from leukocyte adhesion deficiencies, most often resulting in severe recurrent infections and, at the same time, a predisposition for autoimmune diseases. So far, studies on the role of β2 integrins in vivo employed mice with a constitutive knockout of all β2 integrins or either family member, respectively, which complicated the differentiation between the direct and indirect effects of β2 integrin deficiency for distinct cell types. The recent generation and characterization of transgenic mice with a cell-type-specific knockdown of β2 integrins by our group has enabled the dissection of cell-specific roles of β2 integrins. Further, integrin receptors have been recognized as target receptors for the treatment of inflammatory diseases as well as tumor therapy. However, whereas both agonistic and antagonistic agents yielded beneficial effects in animal models, the success of clinical trials was limited in most cases and was associated with unwanted side effects. This unfavorable outcome is most probably related to the systemic effects of the used compounds on all leukocytes, thereby emphasizing the need to develop formulations that target distinct types of leukocytes to modulate β2 integrin activity for therapeutic applications.
Collapse
Affiliation(s)
| | | | | | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (T.K.); (C.H.); (M.B.)
| |
Collapse
|
6
|
Huang S, Chen Y, Gong F, Chen W, Zheng Y, Zhao B, Shi W, Yang Z, Qu H, Mao E, Chen E. Septic macrophages induce T cells immunosuppression in a cell-cell contact manner with the involvement of CR3. Heliyon 2024; 10:e23266. [PMID: 38187232 PMCID: PMC10770445 DOI: 10.1016/j.heliyon.2023.e23266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 01/09/2024] Open
Abstract
Background In addition to excessive inflammation, immunosuppression has been recognized as a contributing factor to poor prognosis of sepsis. Although it has been reported that T cells can become functionally impaired during sepsis, the underlying mechanisms responsible for this phenomenon remain unclear. This study aims to elucidate the mechanisms by which macrophages induce immunosuppression in T cells. Methods In an in vivo setting, C57BL-6J mice were subjected to cecal ligation and puncture (CLP) with or without depletion of macrophages, and the functions of T cells were assessed. In vitro experiments involved direct co-culture or separate culture of T cells and septic macrophages using a transwell system, followed by analysis of T cell immunity. Additionally, a siRNA targeting CD18 on macrophages was utilized to investigate the role of complement receptor 3 (CR3). Results Both macrophages and T cells exhibited immunosuppression during sepsis. In the in vivo experiments, the absence of macrophages partially alleviated T cell immunosuppression, as evidenced by restored vitality, increased production of TNF-α and IFN-γ, elevated CD8+ T cell levels, and decreased CD25+ T cell levels. In the in vitro experiments, direct co-culture of T cells with septic macrophages resulted in diminished T cell immunity, which was improved when T cells and macrophages were separated by a chamber wall. The expression of CR3 (CD11b/CD18) was upregulated on septic macrophages, and silencing of CD18 led to decreased TNF-α production by T cells, reduced CD4+ T cell numbers, and increased CD25+ T cell numbers. Conclusion In sepsis, macrophages induce immunosuppression in T cells through direct cell-cell contact, with the involvement of CR3.
Collapse
Affiliation(s)
- Shunwei Huang
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai, China
| | - Ying Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai, China
| | - Fangchen Gong
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai, China
| | - Weiwei Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai, China
| | - Yanjun Zheng
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai, China
| | - Bing Zhao
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai, China
| | - Wen Shi
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai, China
| | - Zhitao Yang
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai, China
| | - Hongping Qu
- Department of Intensive Care, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai, China
| | - Enqiang Mao
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai, China
| | - Erzhen Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine. Shanghai, China
| |
Collapse
|
7
|
Na K, Oh BC, Jung Y. Multifaceted role of CD14 in innate immunity and tissue homeostasis. Cytokine Growth Factor Rev 2023; 74:100-107. [PMID: 37661484 DOI: 10.1016/j.cytogfr.2023.08.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023]
Abstract
CD14 is a co-receptor of Toll-like receptor (TLR)- 4, with a critical role in innate immune responses. CD14 recognizes bacterial lipopolysaccharides, pathogen-, and damage-associated molecular patterns, thereby facilitating inflammatory immune responses. In addition to its well-established association with TLR4, CD14 is also implicated in TLR4-independent signaling, which leads to the apoptotic death of differentiated dendritic cells and activation of the noncanonical inflammasome pathway. CD14 also has a role beyond that of the immune responses. It contributes to tissue homeostasis by promoting the clearance of various apoptotic cells via recognizing externalized phosphatidylinositol phosphates. CD14 also has context-dependent roles, particularly in barrier tissues that include the skin and gastrointestinal tract. For example, CD14+ dendritic cells in the skin can induce immunostimulatory or immunosuppressive responses. In the gastrointestinal system, CD14 is involved in producing inflammatory cytokines in inflammatory bowel disease and maintaining of intestinal integrity. This review focuses on the multifaceted roles of CD14 in innate immunity and its potential regulatory functions in barrier tissues characterized by rapid cell renewal. By providing insights into the diverse functions of CD14, this review offers potential therapeutic implications for this versatile molecule in immune modulation and tissue homeostasis.
Collapse
Affiliation(s)
- Kunhee Na
- Department of Health Science and Technology, Gachon Advanced Institute for Health Science & Technology, Gachon University, Incheon 21999, the Republic of Korea
| | - Byung-Chul Oh
- Department of Health Science and Technology, Gachon Advanced Institute for Health Science & Technology, Gachon University, Incheon 21999, the Republic of Korea; Department of Physiology, College of Medicine, Gachon University, Incheon 21999, the Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, the Republic of Korea.
| | - YunJae Jung
- Department of Health Science and Technology, Gachon Advanced Institute for Health Science & Technology, Gachon University, Incheon 21999, the Republic of Korea; Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, the Republic of Korea; Department of Microbiology, College of Medicine, Gachon University, Incheon 21999, the Republic of Korea.
| |
Collapse
|
8
|
Ogbodo E, Michelangeli F, Williams JHH. Exogenous heat shock proteins HSPA1A and HSPB1 regulate TNF-α, IL-1β and IL-10 secretion from monocytic cells. FEBS Open Bio 2023; 13:1922-1940. [PMID: 37583307 PMCID: PMC10549225 DOI: 10.1002/2211-5463.13695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/31/2023] [Accepted: 08/14/2023] [Indexed: 08/17/2023] Open
Abstract
Endogenous molecules, such as heat shock proteins (HSP), can function as danger signals when released into the extracellular environment in response to cell stress, where they elicit an immune response such as cytokine secretion. There has also been some suggestion that contamination of exogenous HSPs with lipopolysaccharide (LPS) may be responsible for these effects. This study investigates the effects of exogenous HSPA1A and HSPB1 on the activation of immune cells and the resulting secretion of cytokines, which are involved in inflammatory responses. To address whether exogenous HSPs can directly activate cytokine secretion, naïve U937 cells, differentiated U937 cells and peripheral blood mononuclear cells (PBMCs) were treated with either exogenously applied HSPA1A or HSPB1 and then secreted IL-1β, TNF-α and IL-10 were measured by ELISA. Both HSPs were able to induce a dose-dependent increase in IL-10 secretion from naïve U937 cells and dose-dependent IL-1β, TNF-α and IL-10 secretion were also observed in differentiated U937 cells and PBMCs. We also observed that CD14 affects the secretion levels of IL-1β, TNF-α and IL-10 from cells in response to exogenous HSP treatment. In addition, HSPA1A and HSPB1 were shown to interact with CD14, CD36 and CD11b extracellular receptor proteins. Several approaches used in this study indicate that HSP-induced cytokine secretion is largely independent of any contaminating LPS in the samples.
Collapse
|
9
|
Zubova SV, Kosyakova NI, Grachev SV, Prokhorenko IR. Co-Activation of Human Whole Blood Cells with Lipopolysaccharides and an Allergen. Life (Basel) 2023; 13:1672. [PMID: 37629528 PMCID: PMC10455811 DOI: 10.3390/life13081672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/26/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023] Open
Abstract
The investigation of common inflammation mechanisms caused by exogenic compounds of microbial origin and allergens is one of the most important tasks in current biomedical science. The main manifestations of immune cell activation caused by pro-inflammatory agents are changes in receptor quantity on the surface of immune cells and the production of cytokines and chemokines by blood cells. The levels of expression of TLR4, CD14, and CD11b in the monocytes and neutrophils of human whole blood in response to LPS E. coli, Der p 2 allergen, or their combination reflect different functional activities in these cells, while the composition and amount of produced cytokines reflect the biological activity of the studied agonists. The activity of Der p 2 allergen in ex vivo experiments on whole blood samples is significantly lower compared with its activity in vitro in isolated PBMC cells, which should be taken into account when transferring the results obtained for isolated cells to whole blood cells. LPS R. capsulatus PG significantly decreases the synthesis of MyD88-dependent NF-κB-regulated cytokines activated by LPS E. coli, Der p 2, or their combination. This indirectly indicates the general mechanisms of cell activation caused by these structures and the unified mechanism of the protective action of LPS R. capsulatus PG against both endotoxin and a combination of endotoxin and the allergen.
Collapse
Affiliation(s)
- Svetlana V. Zubova
- Hospital of Pushchino Scientific Center, Russian Academy of Sciences, Pushchino 142290, Russia;
- Department of Molecular Biomedicine, Institute of Basic Biological Problems, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino 142290, Russia; (S.V.G.); (I.R.P.)
| | - Ninel I. Kosyakova
- Hospital of Pushchino Scientific Center, Russian Academy of Sciences, Pushchino 142290, Russia;
| | - Sergey V. Grachev
- Department of Molecular Biomedicine, Institute of Basic Biological Problems, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino 142290, Russia; (S.V.G.); (I.R.P.)
| | - Isabella R. Prokhorenko
- Department of Molecular Biomedicine, Institute of Basic Biological Problems, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Pushchino 142290, Russia; (S.V.G.); (I.R.P.)
| |
Collapse
|
10
|
Choconta JL, Labi V, Dumbraveanu C, Kalpachidou T, Kummer KK, Kress M. Age-related neuroimmune signatures in dorsal root ganglia of a Fabry disease mouse model. Immun Ageing 2023; 20:22. [PMID: 37173694 PMCID: PMC10176851 DOI: 10.1186/s12979-023-00346-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023]
Abstract
Pain in Fabry disease (FD) is generally accepted to result from neuronal damage in the peripheral nervous system as a consequence of excess lipid storage caused by alpha-galactosidase A (α-Gal A) deficiency. Signatures of pain arising from nerve injuries are generally associated with changes of number, location and phenotypes of immune cells within dorsal root ganglia (DRG). However, the neuroimmune processes in the DRG linked to accumulating glycosphingolipids in Fabry disease are insufficiently understood.Therefore, using indirect immune fluorescence microscopy, transmigration assays and FACS together with transcriptomic signatures associated with immune processes, we assessed age-dependent neuroimmune alterations in DRG obtained from mice with a global depletion of α-Gal A as a valid mouse model for FD. Macrophage numbers in the DRG of FD mice were unaltered, and BV-2 cells as a model for monocytic cells did not show augmented migratory reactions to glycosphingolipids exposure suggesting that these do not act as chemoattractants in FD. However, we found pronounced alterations of lysosomal signatures in sensory neurons and of macrophage morphology and phenotypes in FD DRG. Macrophages exhibited reduced morphological complexity indicated by a smaller number of ramifications and more rounded shape, which were age dependent and indicative of premature monocytic aging together with upregulated expression of markers CD68 and CD163.In our FD mouse model, the observed phenotypic changes in myeloid cell populations of the DRG suggest enhanced phagocytic and unaltered proliferative capacity of macrophages as compared to wildtype control mice. We suggest that macrophages may participate in FD pathogenesis and targeting macrophages at an early stage of FD may offer new treatment options other than enzyme replacement therapy.
Collapse
Affiliation(s)
- Jeiny Luna Choconta
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Labi
- Institute of Developmental Immunology, Medical University of Innsbruck, Innsbruck, Austria
| | | | | | - Kai K Kummer
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
11
|
Zhang Q, Zhang S, Chen J, Xie Z. The Interplay between Integrins and Immune Cells as a Regulator in Cancer Immunology. Int J Mol Sci 2023; 24:6170. [PMID: 37047140 PMCID: PMC10093897 DOI: 10.3390/ijms24076170] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Integrins are a group of heterodimers consisting of α and β subunits that mediate a variety of physiological activities of immune cells, including cell migration, adhesion, proliferation, survival, and immunotolerance. Multiple types of integrins act differently on the same immune cells, while the same integrin may exert various effects on different immune cells. In the development of cancer, integrins are involved in the regulation of cancer cell proliferation, invasion, migration, and angiogenesis; conversely, integrins promote immune cell aggregation to mediate the elimination of tumors. The important roles of integrins in cancer progression have provided valuable clues for the diagnosis and targeted treatment of cancer. Furthermore, many integrin inhibitors have been investigated in clinical trials to explore effective regimens and reduce side effects. Due to the complexity of the mechanism of integrin-mediated cancer progression, challenges remain in the research and development of cancer immunotherapies (CITs). This review enumerates the effects of integrins on four types of immune cells and the potential mechanisms involved in the progression of cancer, which will provide ideas for more optimal CIT in the future.
Collapse
Affiliation(s)
- Qingfang Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Shuo Zhang
- College of Basic Medical, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Jianrui Chen
- College of Basic Medical, Nanchang University, Nanchang 330006, China
- Queen Mary School, Medical Department, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- College of Basic Medical, Nanchang University, Nanchang 330006, China
| |
Collapse
|
12
|
Yazar V, Yilmaz IC, Bulbul A, Klinman DM, Gursel I. Gene network landscape of mouse splenocytes reveals integrin complex as the A151 ODN-responsive hub molecule in the immune transcriptome. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:553-565. [PMID: 36895952 PMCID: PMC9989320 DOI: 10.1016/j.omtn.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Homeostatic restoration of an inflammatory response requires quenching of the immune system after pathogen threats vanish. A continued assault orchestrated by host defense results in tissue destruction or autoimmunity. A151 is the epitome of synthetic oligodeoxynucleotides (ODNs) that curb the immune response by a subset of white corpuscles through repetitive telomere-derived TTAGGG sequences. Currently, the genuine effect of A151 on the immune cell transcriptome remains unknown. Here, we leveraged an integrative approach where weighted gene co-expression network analysis (WGCNA), differential gene expression analysis, and gene set enrichment analysis (GSEA) of our in-house microarray datasets aided our understanding of how A151 ODN suppresses the immune response in mouse splenocytes. Our bioinformatics results, together with experimental validations, indicated that A151 ODN acts on components of integrin complexes, Itgam and Itga6, to interfere with immune cell adhesion and thereby suppresses the immune response in mice. Moreover, independent lines of evidence in this work converged on the observation that cell adhesion by integrin complexes serves as a focal point for cellular response to A151 ODN treatment in immune cells. Taken together, the outcome of this study sheds light on the molecular basis of immune suppression by a clinically useful DNA-based therapeutic agent.
Collapse
Affiliation(s)
- Volkan Yazar
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Thorlab, Molecular Biology and Genetics Department, Faculty of Science, Ihsan Dogramaci Bilkent University, 06800 Ankara, Turkey
| | - Ismail Cem Yilmaz
- Thorlab, Molecular Biology and Genetics Department, Faculty of Science, Ihsan Dogramaci Bilkent University, 06800 Ankara, Turkey
- Izmir Biomedicine and Genome Center, Dokuz Eylul University, Balcova, Izmir, Turkey
| | - Artun Bulbul
- Thorlab, Molecular Biology and Genetics Department, Faculty of Science, Ihsan Dogramaci Bilkent University, 06800 Ankara, Turkey
| | - Dennis M. Klinman
- Immune Modulation Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Ihsan Gursel
- Thorlab, Molecular Biology and Genetics Department, Faculty of Science, Ihsan Dogramaci Bilkent University, 06800 Ankara, Turkey
- Izmir Biomedicine and Genome Center, Dokuz Eylul University, Balcova, Izmir, Turkey
- Corresponding author: Ihsan Gursel, PhD, Izmir Biomedicine and Genome Center, Dokuz Eylul University, Balcova, Izmir, Turkey.
| |
Collapse
|
13
|
Hou L, Liu J, Sun F, Huang R, Chang R, Ruan Z, Wang Y, Zhao J, Wang Q. Integrin Mac1 mediates paraquat and maneb-induced learning and memory impairments in mice through NADPH oxidase-NLRP3 inflammasome axis-dependent microglial activation. J Neuroinflammation 2023; 20:42. [PMID: 36804009 PMCID: PMC9938991 DOI: 10.1186/s12974-023-02732-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 02/13/2023] [Indexed: 02/20/2023] Open
Abstract
INTRODUCTION The mechanisms of cognitive impairments in Parkinson's disease (PD) remain unknown. Accumulating evidence revealed that brain neuroinflammatory response mediated by microglial cells contributes to cognitive deficits in neuropathological conditions and macrophage antigen complex-1 (Mac1) is a key factor in controlling microglial activation. OBJECTIVES To explore whether Mac1-mediated microglial activation participates in cognitive dysfunction in PD using paraquat and maneb-generated mouse PD model. METHODS Cognitive performance was measured in wild type and Mac1-/- mice using Morris water maze test. The role and mechanisms of NADPH oxidase (NOX)-NLRP3 inflammasome axis in Mac1-mediated microglial dysfunction, neuronal damage, synaptic degeneration and phosphorylation (Ser129) of α-synuclein were explored by immunohistochemistry, Western blot and RT-PCR. RESULTS Genetic deletion of Mac1 significantly ameliorated learning and memory impairments, neuronal damage, synaptic loss and α-synuclein phosphorylation (Ser129) caused by paraquat and maneb in mice. Subsequently, blocking Mac1 activation was found to mitigate paraquat and maneb-elicited microglial NLRP3 inflammasome activation in both in vivo and in vitro. Interestingly, stimulating activation of NOX by phorbol myristate acetate abolished the inhibitory effects of Mac1 blocking peptide RGD on paraquat and maneb-provoked NLRP3 inflammasome activation, indicating a key role of NOX in Mac1-mediated NLRP3 inflammasome activation. Furthermore, NOX1 and NOX2, two members of NOX family, and downstream PAK1 and MAPK pathways were recognized to be essential for NOX to regulate NLRP3 inflammasome activation. Finally, a NLRP3 inflammasome inhibitor glybenclamide abrogated microglial M1 activation, neurodegeneration and phosphorylation (Ser129) of α-synuclein elicited by paraquat and maneb, which were accompanied by improved cognitive capacity in mice. CONCLUSIONS Mac1 was involved in cognitive dysfunction in a mouse PD model through NOX-NLRP3 inflammasome axis-dependent microglial activation, providing a novel mechanistic basis of cognitive decline in PD.
Collapse
Affiliation(s)
- Liyan Hou
- grid.411971.b0000 0000 9558 1426Dalian Medical University Library, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China ,grid.411971.b0000 0000 9558 1426National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044 China
| | - Jianing Liu
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Fuqiang Sun
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Ruixue Huang
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Rui Chang
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Zhengzheng Ruan
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Ying Wang
- grid.411971.b0000 0000 9558 1426School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044 China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China.
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China. .,School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China.
| |
Collapse
|
14
|
Pant H, Hercus TR, Tumes DJ, Yip KH, Parker MW, Owczarek CM, Lopez AF, Huston DP. Translating the biology of β common receptor-engaging cytokines into clinical medicine. J Allergy Clin Immunol 2023; 151:324-344. [PMID: 36424209 DOI: 10.1016/j.jaci.2022.09.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/16/2022] [Accepted: 09/29/2022] [Indexed: 11/23/2022]
Abstract
The family of cytokines that comprises IL-3, IL-5, and GM-CSF was discovered over 30 years ago, and their biological activities and resulting impact in clinical medicine has continued to expand ever since. Originally identified as bone marrow growth factors capable of acting on hemopoietic progenitor cells to induce their proliferation and differentiation into mature blood cells, these cytokines are also recognized as key mediators of inflammation and the pathobiology of diverse immunologic diseases. This increased understanding of the functional repertoire of IL-3, IL-5, and GM-CSF has led to an explosion of interest in modulating their functions for clinical management. Key to the successful clinical translation of this knowledge is the recognition that these cytokines act by engaging distinct dimeric receptors and that they share a common signaling subunit called β-common or βc. The structural determination of how IL-3, IL-5, and GM-CSF interact with their receptors and linking this to their differential biological functions on effector cells has unveiled new paradigms of cell signaling. This knowledge has paved the way for novel mAbs and other molecules as selective or pan inhibitors for use in different clinical settings.
Collapse
Affiliation(s)
- Harshita Pant
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia; Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Timothy R Hercus
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
| | - Damon J Tumes
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
| | - Kwok Ho Yip
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia
| | - Michael W Parker
- Bio 21 Institute, The University of Melbourne, Melbourne, Australia; St Vincent's Institute of Medical Research, Melbourne, Australia
| | | | - Angel F Lopez
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, Australia; Adelaide Medical School, University of Adelaide, Adelaide, Australia.
| | - David P Huston
- Texas A&M University School of Medicine, Houston, Tex; Houston Methodist Hospital and Research Institute, Houston, Tex.
| |
Collapse
|
15
|
Zhou K, Yuan L, Liu H, Du X, Yao Y, Qin L, Yang M, Xu K, Wu X, Wang L, Xiang Y, Qu X, Qin X, Liu C. ITGB4 deficiency in airway epithelia enhances HDM-induced airway inflammation through hyperactivation of TLR4 signaling pathway. J Leukoc Biol 2023; 113:216-227. [PMID: 36822178 DOI: 10.1093/jleuko/qiac013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Indexed: 01/18/2023] Open
Abstract
Airway epithelial cells (AECs) are the first cell barrier of the respiratory system against external stimuli that play a critical role in the development of asthma. It is known that AECs play a key role in asthma susceptibility and severity. ITGB4 is a downregulated adhesion molecule in the airway epithelia of asthma patients, which was involved in the exaggerated lung inflammation after allergy stimulation. Toll-like receptor 4 (TLR4) in AECs has also been shown to play a crucial role in the development of lung inflammation in asthma patients. However, the specific intrinsic regulatory mechanism of TLR4 in AECs are still obscure. In this article, we demonstrated that ITGB4 deficiency in AECs enhances HDM-induced airway inflammation through hyperactivation of the TLR4 signaling pathway, which is mediated by inhibition of FYN phosphorylation. Moreover, TLR4-antagonist treatment or blockade of FYN can inhibit or exaggerate lung inflammation in HDM-stressed ITGB4-deficient mice, separately. Together, these results demonstrated that ITGB4 deficiency in AECs enhances HDM-induced lung inflammatory response through the ITGB4-FYN-TLR4 axis, which may provide new therapeutic approaches for the management of lung inflammation in asthma.
Collapse
Affiliation(s)
- Kai Zhou
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
- Department of Physiology, School of Basic Medicine Science, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
- Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| | - Lin Yuan
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
- Department of Physiology, School of Basic Medicine Science, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
- Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| | - Xizi Du
- Department of Physiology, School of Basic Medicine Science, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| | - Ye Yao
- Department of Physiology, School of Basic Medicine Science, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| | - Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
- Basic and Clinical Research Laboratory of Major Respiratory Diseases, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| | - Ming Yang
- Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Elizabeth Street, Callaghan, New South Wales 2892921, Australia
| | - Kun Xu
- School of Public Health, Jilin University, Xinmin Dajie Street, Changchun 130000, China
| | - Xinyu Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| | - Leyuan Wang
- Department of Physiology, School of Basic Medicine Science, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine Science, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| | - Chi Liu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
- Department of Physiology, School of Basic Medicine Science, Central South University, Xiangya Road Street, Changsha, Hunan 410078, China
| |
Collapse
|
16
|
Controlling Macrophage Polarization to Modulate Inflammatory Cues Using Immune-Switch Nanoparticles. Int J Mol Sci 2022; 23:ijms232315125. [PMID: 36499452 PMCID: PMC9739781 DOI: 10.3390/ijms232315125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
The persistence of inflammatory mediators in tissue niches significantly impacts regenerative outcomes and contributes to chronic diseases. Interleukin-4 (IL4) boosts pro-healing phenotypes in macrophages (Mφ) and triggers the activation of signal transducer and activator of transcription 6 (STAT6). Since the IL4/STAT6 pathway reduces Mφ responsiveness to inflammation in a targeted and precise manner, IL4 delivery offers personalized possibilities to overcome inflammatory events. Despite its therapeutic potential, the limited success of IL4-targeted delivery is hampered by inefficient vehicles. Magnetically assisted technologies offer precise and tunable nanodevices for the delivery of cytokines by combining contactless modulation, high tissue penetration, imaging features, and low interference with the biological environment. Although superparamagnetic iron oxide nanoparticles (SPION) have shown clinical applicability in imaging, SPION-based approaches have rarely been explored for targeted delivery and cell programming. Herein, we hypothesized that SPION-based carriers assist in efficient IL4 delivery to Mφ, favoring a pro-regenerative phenotype (M2φ). Our results confirmed the efficiency of SPION-IL4 and Mφ responsiveness to SPION-IL4 with evidence of STAT6-mediated polarization. SPION-IL4-treated Mφ showed increased expression of M2φ associated-mediators (IL10, ARG1, CCL2, IL1Ra) when compared to the well-established soluble IL4. The ability of SPION-IL4 to direct Mφ polarization using sophisticated magnetic nanotools is valuable for resolving inflammation and assisting innovative strategies for chronic inflammatory conditions.
Collapse
|
17
|
Gopalakrishnan A, Richard K, Wahid R, Harley R, Sztein MB, Hawkins LD, Vogel SN. E6020, a TLR4 Agonist Adjuvant, Enhances Both Antibody Titers and Isotype Switching in Response to Immunization with Hapten-Protein Antigens and Is Diminished in Mice with TLR4 Signaling Insufficiency. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1950-1959. [PMID: 36426935 PMCID: PMC9643654 DOI: 10.4049/jimmunol.2200495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/14/2022] [Indexed: 12/30/2022]
Abstract
The mechanisms by which TLR4-based adjuvants enhance immunogenicity are not fully understood. We have taken advantage of a novel knock-in mouse strain that homozygously expresses two single-nucleotide polymorphisms (SNPs) that are homologous to human TLR4 (rs4986790 and rs4986791) and have been associated with LPS hyporesponsiveness in vivo and in vitro. TLR4-SNP (coexpressing mutations D298G/N397I in TLR4) mice that recapitulate the human phenotype were compared with wild-type (WT) mice for their hapten-specific Ab responses after immunization with hapten 4-hydroxy-3-nitrophenyl acetyl (NP) NP-Ficoll or NP-OVA in the absence or presence of a water-soluble TLR4 analog adjuvant, E6020. IgM and IgG anti-NP responses were comparable in WT and TLR4-SNP mice after immunization with either NP-Ficoll or NP-OVA only. E6020 significantly yet transiently improved the IgM and IgG anti-NP responses of both WT and TLR4-SNP mice to NP-Ficoll (T-independent), with modestly enhanced Ab production in WT mice. In contrast, T-dependent (NP-OVA), adjuvant-enhanced responses showed sustained elevation of NP-specific Ab titers in WT mice, intermediate responses in TLR4-SNP mice, and negligible enhancement in TLR4-/- mice. E6020-enhanced early humoral responses in WT and TLR4-SNP mice to NP-OVA favored an IgG1 response. After a second immunization, however, the immune responses of TLR4-SNP mice remained IgG1 dominant, whereas WT mice reimmunized with NP-OVA and E6020 exhibited increased anti-NP IgG2c titers and a sustained increase in the IgG1 and IgG2c production by splenocytes. These findings indicate that E6020 increases and sustains Ab titers and promotes isotype class switching, as evidenced by reduced titers and IgG1-dominant immune responses in mice with TLR4 insufficiency.
Collapse
Affiliation(s)
- Archana Gopalakrishnan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD
| | - Katharina Richard
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD
| | - Rezwanul Wahid
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD
| | - Regina Harley
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD
| | | | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
18
|
Wang S, Chen Y, Ling Z, Li J, Hu J, He F, Chen Q. The role of dendritic cells in the immunomodulation to implanted biomaterials. Int J Oral Sci 2022; 14:52. [PMCID: PMC9636170 DOI: 10.1038/s41368-022-00203-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022] Open
Abstract
Considering the substantial role played by dendritic cells (DCs) in the immune system to bridge innate and adaptive immunity, studies on DC-mediated immunity toward biomaterials principally center on their adjuvant effects in facilitating the adaptive immunity of codelivered antigens. However, the effect of the intrinsic properties of biomaterials on dendritic cells has not been clarified. Recently, researchers have begun to investigate and found that biomaterials that are nonadjuvant could also regulate the immune function of DCs and thus affect subsequent tissue regeneration. In the case of proteins adsorbed onto biomaterial surfaces, their intrinsic properties can direct their orientation and conformation, forming “biomaterial-associated molecular patterns (BAMPs)”. Thus, in this review, we focused on the intrinsic physiochemical properties of biomaterials in the absence of antigens that affect DC immune function and summarized the underlying signaling pathways. Moreover, we preliminarily clarified the specific composition of BAMPs and the interplay between some key molecules and DCs, such as heat shock proteins (HSPs) and high mobility group box 1 (HMGB1). This review provides a new direction for future biomaterial design, through which modulation of host immune responses is applicable to tissue engineering and immunotherapy.
Collapse
Affiliation(s)
- Siyuan Wang
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Yanqi Chen
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Zhaoting Ling
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Jia Li
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Jun Hu
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Fuming He
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| | - Qianming Chen
- grid.13402.340000 0004 1759 700XStomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006 China
| |
Collapse
|
19
|
Cao M, Ma L, Yan C, Wang H, Ran M, Chen Y, Wang X, Liang X, Chai L, Li X. Mouse Ocilrp2/Clec2i negatively regulates LPS-mediated IL-6 production by blocking Dap12-Syk interaction in macrophage. Front Immunol 2022; 13:984520. [PMID: 36300111 PMCID: PMC9589251 DOI: 10.3389/fimmu.2022.984520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/20/2022] [Indexed: 11/24/2022] Open
Abstract
C-type lectin Ocilrp2/Clec2i is widely expressed in dendritic cells, lymphokine-activated killer cells and activated T cells. Previous studies have shown that Ocilrp2 is an important regulator in the activation of T cells and NK cells. However, the role of Ocilrp2 in the inflammatory responses by activated macrophages is currently unknown. This study investigated the expression of inflammatory cytokines in LPS-induced macrophages from primary peritoneal macrophages silenced by specific siRNA target Ocilrp2. Ocilrp2 was significantly downregulated in macrophages via NF-κB and pathways upon LPS stimuli or VSV infection. Silencing Ocilrp2 resulted in the increased expression of IL-6 in LPS-stimulated peritoneal macrophages and mice. Moreover, IL-6 expression was reduced in LPS-induced Ocilrp2 over-expressing iBMDM cells. Furthermore, we found that Ocilrp2-related Syk activation is responsible for expressing inflammatory cytokines in LPS-stimulated macrophages. Silencing Ocilrp2 significantly promotes the binding of Syk to Dap12. Altogether, we identified the Ocilrp2 as a critical role in the TLR4 signaling pathway and inflammatory macrophages’ immune regulation, and added mechanistic insights into the crosstalk between TLR and Syk signaling.
Collapse
Affiliation(s)
- Mingya Cao
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
- Institute of Translational Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Lina Ma
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Chenyang Yan
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Han Wang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Mengzhe Ran
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Ying Chen
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Xiao Wang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Xiaonan Liang
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
| | - Lihui Chai
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
- Institute of Translational Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
- *Correspondence: Lihui Chai, ; Xia Li,
| | - Xia Li
- Joint National Laboratory for Antibody Drug Engineering, The First Affiliated Hospital, School of Medicine, Henan University, Kaifeng, China
- Institute of Translational Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
- *Correspondence: Lihui Chai, ; Xia Li,
| |
Collapse
|
20
|
Tan YY, Yue SR, Lu AP, Zhang L, Ji G, Liu BC, Wang RR. The improvement of nonalcoholic steatohepatitis by Poria cocos polysaccharides associated with gut microbiota and NF-κB/CCL3/CCR1 axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 103:154208. [PMID: 35691078 DOI: 10.1016/j.phymed.2022.154208] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH) has been linked to inflammation induced by intestinal microbiota. Poria cocos polysaccharides (PCP) possesses anti-inflammation and immunomodulation functions; however, its preventive effects against NASH and potential mechanisms need to be explored. METHODS The composition of PCP was determined using ion chromatography. C57BL/6 mice were administered the methionine and choline deficient (MCD) diet for 4 weeks to establish the NASH model or methionine-choline-sufficient (MCS) diet to serve as the control. Mice were assigned to the MCS group, MCD group, low-dose PCP (LP) group, and high-dose PCP (HP) group, and were administered the corresponding medications via gavage. Serum biochemical index analysis and liver histopathology examination were performed to verify the successful establishment of NASH model and to evaluate the efficacy of PCP. The composition of intestinal bacteria was profiled through 16S rRNA gene sequencing. Hepatic RNA sequencing (RNA-Seq) was performed to explore the potential mechanisms, which were further confirmed using qPCR, western blot, and immunohistochemistry. RESULTS PCP consists of glucose, galactose, mannose, D-glucosamine hydrochloride, xylose, arabinose, and fucose. PCP could significantly alleviate symptoms of NASH, including histological liver damage, impaired hepatic function, and increased oxidative stress. Meanwhile, HP could reshape the composition of intestinal bacteria by significantly increasing the relative abundance of Faecalibaculum and decreasing the level of endotoxin load derived from gut bacteria. PCP could also downregulate the expression of pathways associated with immunity and inflammation, including the chemokine signaling pathway, Toll-like receptor signaling pathway, and NF-kappa B signaling pathway. The expression levels of CCL3 and CCR1 (involved in the chemokine signaling pathway), Tlr4, Cd11b, and NF-κb (involved in the NF-kappa B signaling pathway), and Tnf-α (involved in the TNF signaling pathway) were significantly reduced in the HP group compared to the MCD group. CONCLUSIONS PCP could prevent the development of NASH, which may be associated with the modulation of intestinal microbiota and the downregulation of the NF-κB/CCL3/CCR1 axis.
Collapse
Affiliation(s)
- Yi-Yun Tan
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Si-Ran Yue
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ai-Ping Lu
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Lei Zhang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guang Ji
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Bao-Cheng Liu
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Rui-Rui Wang
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
21
|
Frings VG, Jopp L, Srivastava M, Presser D, Goebeler M, Schmidt M. Stress signaling and STAT1 activation characterize the keratinocytic gene expression pattern in Hidradenitis suppurativa. J Eur Acad Dermatol Venereol 2022; 36:2488-2498. [PMID: 35881108 DOI: 10.1111/jdv.18465] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/11/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The pathogenetic factors generating the innate immune signal necessary for T cell activation, initiation and chronification of Hidradenitis suppurativa (HS, also known as Acne inversa) are still poorly understood. Emerging evidence suggests that a defective keratinocyte function critically contributes to HS disease development and progression. OBJECTIVES To elucidate the role of keratinocytes in HS lesion formation, we compared the transcriptomes of lesional and perilesional epidermis isolated from HS patients by RNA sequencing (RNA Seq). METHODS Pairwise-matched lesional and perilesional HS skin samples of five different donors were obtained and epidermal keratinocytes freshly isolated and processed for RNA extraction and RNA seq. Lesionally regulated genes were analyzed by large scale promotor analysis and functional annotation clustering to identify epidermally overrepresented transcription factor binding sites and functionally related gene groups. Results were experimentally validated with independent epidermal isolates of patient-matched lesional and perilesional HS skin employing qRT-PCR, cell culture, immunoblot, and immunostaining. RESULTS We show that HS is characterized by a strong epidermal stress state evident by a significant overrepresentation of an AP-1-driven gene signature and a substantial activation of the stress-activated cJun N-terminal kinase (JNK) pathway in lesional epidermis. Additionally, our data reveal a strong induction of STAT1 activation in lesional HS epidermis that likely results from IFNγ production and triggered expression of key inflammatory genes coordinating innate immune activation and the adaptive T cell response in HS. CONCLUSIONS Our data implicate a key role of stress signaling and JAK/STAT1 activation in disease progression of HS and suggest interference with JAK/STAT1 signaling as a potentially promising therapeutic approach for HS.
Collapse
Affiliation(s)
- V G Frings
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Germany
| | - L Jopp
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Germany
| | - M Srivastava
- Core Unit Systemmedizin (SysMed), Medical Faculty, University of Würzburg, Würzburg, Germany
| | - D Presser
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Germany
| | - M Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Germany
| | - M Schmidt
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Germany
| |
Collapse
|
22
|
Mzyk P, Hernandez H, Le T, Ramirez JR, McDowell CM. Toll-Like Receptor 4 Signaling in the Trabecular Meshwork. Front Cell Dev Biol 2022; 10:936115. [PMID: 35912101 PMCID: PMC9335276 DOI: 10.3389/fcell.2022.936115] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/21/2022] [Indexed: 12/04/2022] Open
Abstract
Primary open-angle glaucoma is one of the leading causes of blindness worldwide. With limited therapeutics targeting the pathogenesis at the trabecular meshwork (TM), there is a great need for identifying potential new targets. Recent evidence has implicated Toll-like receptor 4 (TLR4) and it is signaling pathway in augmenting the effects of transforming growth factor beta-2 (TGFβ2) and downstream extracellular matrix production. In this review, we examine the role of TLR4 signaling in the trabecular meshwork and the interplay between endogenous activators of TLR4 (damage-associated molecular patterns (DAMPs)), extracellular matrix (ECM), and the effect on intraocular pressure.
Collapse
Affiliation(s)
- Philip Mzyk
- University of Wisconsin-Madison, Madison, WI, United States
| | | | - Thanh Le
- University of Houston-Victoria, Victoria, TX, United States
| | | | | |
Collapse
|
23
|
Bednarczyk M, Bolduan V, Haist M, Stege H, Hieber C, Johann L, Schelmbauer C, Blanfeld M, Karram K, Schunke J, Klaus T, Tubbe I, Montermann E, Röhrig N, Hartmann M, Schlosser J, Bopp T, Clausen BE, Waisman A, Bros M, Grabbe S. β2 Integrins on Dendritic Cells Modulate Cytokine Signaling and Inflammation-Associated Gene Expression, and Are Required for Induction of Autoimmune Encephalomyelitis. Cells 2022; 11:cells11142188. [PMID: 35883631 PMCID: PMC9322999 DOI: 10.3390/cells11142188] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 01/27/2023] Open
Abstract
Heterodimeric β2 integrin surface receptors (CD11a-d/CD18) are specifically expressed by leukocytes that contribute to pathogen uptake, cell migration, immunological synapse formation and cell signaling. In humans, the loss of CD18 expression results in leukocyte adhesion deficiency syndrome (LAD-)1, largely characterized by recurrent severe infections. All available mouse models display the constitutive and ubiquitous knockout of either α or the common β2 (CD18) subunit, which hampers the analysis of the cell type-specific role of β2 integrins in vivo. To overcome this limitation, we generated a CD18 gene floxed mouse strain. Offspring generated from crossing with CD11c-Cre mice displayed the efficient knockdown of β2 integrins, specifically in dendritic cells (DCs). Stimulated β2-integrin-deficient splenic DCs showed enhanced cytokine production and the concomitantly elevated activity of signal transducers and activators of transcription (STAT) 1, 3 and 5, as well as the impaired expression of suppressor of cytokine signaling (SOCS) 2–6 as assessed in bone marrow-derived (BM) DCs. Paradoxically, these BMDCs also showed the attenuated expression of genes involved in inflammatory signaling. In line, in experimental autoimmune encephalomyelitis mice with a conditional DC-specific β2 integrin knockdown presented with a delayed onset and milder course of disease, associated with lower frequencies of T helper cell populations (Th)1/Th17 in the inflamed spinal cord. Altogether, our mouse model may prove to be a valuable tool to study the leukocyte-specific functions of β2 integrins in vivo.
Collapse
Affiliation(s)
- Monika Bednarczyk
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Vanessa Bolduan
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Maximilian Haist
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Henner Stege
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Christoph Hieber
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Lisa Johann
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
| | - Carsten Schelmbauer
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
| | - Michaela Blanfeld
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
| | - Khalad Karram
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Jenny Schunke
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Tanja Klaus
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Ingrid Tubbe
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Evelyn Montermann
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Nadine Röhrig
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Maike Hartmann
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Jana Schlosser
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
| | - Tobias Bopp
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
- Institute of Immunology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (L.J.); (C.S.); (M.B.); (K.K.); (B.E.C.); (A.W.)
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Matthias Bros
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (V.B.); (M.H.); (H.S.); (C.H.); (J.S.); (T.K.); (I.T.); (E.M.); (N.R.); (M.H.); (J.S.); (M.B.)
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University of Mainz, Langenbeckstraße 1, 55131 Mainz, Germany;
- Correspondence: ; Tel.: +49-61-3117-4412
| |
Collapse
|
24
|
Eslami-Kaliji F, Mirahmadi-Zare SZ, Nazem S, Shafie N, Ghaedi R, Asadian-Esfahani MH. A label-free SPR biosensor for specific detection of TLR4 expression; introducing of 10-HDA as an antagonist. Int J Biol Macromol 2022; 217:142-149. [PMID: 35817233 DOI: 10.1016/j.ijbiomac.2022.07.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/15/2022] [Accepted: 07/05/2022] [Indexed: 11/05/2022]
Abstract
Toll-like receptor 4 (TLR4) is actively involved in many health-related problems, including transplantation rejection and autoimmune diseases. Therefore, it is important to identify an antagonist to inhibit the TLR4-induced immune cell activation. In our previous study, 10-hydroxy-2-decanoic acid (10-HDA) was introduced as a potential antagonist for TLR4; however, possible interaction between 10-HDA and TLR4 needed to be detected. Due to the ability of surface plasmon resonance (SPR) biosensor to confirm the specific interactions between receptors and ligands, a new configuration of SPR biosensor proposed to detect the possible interaction between 10-HDA and TLR4. Hence, 10-HDA was immobilized using the (3-aminopropyl) triethoxysilane (APTES) polymer as a crosslinking agent on the Ag-MgF2 surface. Besides, genetically modified HEK293T cells with high TLR4 expression were used to study the possible interaction between 10-HDA and TLR4. Surprisingly, the SPR angle was significantly reduced in the presence of HEK cells expressing TLR4, while HEK cells without TLR4 did not affect the SPR angle. So, the proposed SPR biosensor successfully detected the interaction betweenTLR4 and 10-HDA. The sensitivity and detection limit of the biosensor were achieved at 0.05 and 0.5 million cells expressing TLR4, respectively, with a two-fold dynamic range.
Collapse
Affiliation(s)
- Farshid Eslami-Kaliji
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran
| | - Seyede Zohreh Mirahmadi-Zare
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran.
| | - Saeid Nazem
- Department of Physics, Faculty of Science, University of Isfahan, Isfahan, Iran
| | - Negar Shafie
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran
| | - Rassoul Ghaedi
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, 8159358686 Isfahan, Iran
| | | |
Collapse
|
25
|
Villanueva V, Li X, Jimenez V, Faridi HM, Gupta V. CD11b agonists offer a novel approach for treating lupus nephritis. Transl Res 2022; 245:41-54. [PMID: 35288363 PMCID: PMC9167730 DOI: 10.1016/j.trsl.2022.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/20/2022]
Abstract
Lupus nephritis (LN) develops in more than a third of all systemic lupus erythematosus (SLE) patients and is the strongest predictor of morbidity and mortality. Increased circulating levels of type I interferon (IFN I) and anti-double stranded DNA (anti-dsDNA) and anti-RNA binding protein (anti-RNP) antibodies lead to increased glomerular injury via leukocyte activation and glomerular infiltration. Uncontrolled Toll-like receptor (TLR) signaling in leukocytes results in increased production of IFN I and anti-dsDNA antibodies. ITGAM gene codes for integrin CD11b, the α-chain of integrin heterodimer CD11b/CD18, that is highly expressed in leukocytes and modulates TLR-dependent pro-inflammatory signaling. Three nonsynonymous SNPs in the ITGAM gene strongly correlate with increased risk for SLE and LN and with IFN I levels. Here we review the literature on the role of CD11b on leukocytes in LN. We also incorporate conclusions from several recent studies that show that these ITGAM SNPs result in a CD11b protein that is less able to suppress TLR-dependent pro-inflammatory pathways in leukocytes, that activation of CD11b via novel small molecule agonists suppresses TLR-dependent pathways, including reductions in circulating levels of IFN I and anti-dsDNA antibodies, and that CD11b activation reduces LN in model systems. Recent data strongly suggest that integrin CD11b is an exciting new therapeutic target in SLE and LN and that allosteric activation of CD11b is a novel therapeutic paradigm for effectively treating such autoimmune diseases.
Collapse
Affiliation(s)
- Veronica Villanueva
- Drug Discovery Center, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Xiaobo Li
- Drug Discovery Center, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Viviana Jimenez
- Drug Discovery Center, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Hafeez M Faridi
- Department of Pharmaceutical Sciences, College of Pharmacy, Chicago State University, Chicago, Illinois
| | - Vineet Gupta
- Drug Discovery Center, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois.
| |
Collapse
|
26
|
Haist M, Ries F, Gunzer M, Bednarczyk M, Siegel E, Kuske M, Grabbe S, Radsak M, Bros M, Teschner D. Neutrophil-Specific Knockdown of β2 Integrins Impairs Antifungal Effector Functions and Aggravates the Course of Invasive Pulmonal Aspergillosis. Front Immunol 2022; 13:823121. [PMID: 35734179 PMCID: PMC9207500 DOI: 10.3389/fimmu.2022.823121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
β2-integrins are heterodimeric surface receptors that are expressed specifically by leukocytes and consist of a variable α (CD11a-d) and a common β-subunit (CD18). Functional impairment of CD18, which causes leukocyte adhesion deficiency type-1 results in an immunocompromised state characterized by severe infections, such as invasive pulmonary aspergillosis (IPA). The underlying immune defects have largely been attributed to an impaired migratory and phagocytic activity of polymorphonuclear granulocytes (PMN). However, the exact contribution of β2-integrins for PMN functions in-vivo has not been elucidated yet, since the mouse models available so far display a constitutive CD18 knockout (CD18-/- or CD18hypo). To determine the PMN-specific role of β2-integrins for innate effector functions and pathogen control, we generated a mouse line with a Ly6G-specific knockdown of the common β-subunit (CD18Ly6G cKO). We characterized CD18Ly6G cKO mice in-vitro to confirm the PMN-specific knockdown of β2-integrins. Next, we investigated the clinical course of IPA in A. fumigatus infected CD18Ly6G cKO mice with regard to the fungal burden, pulmonary inflammation and PMN response towards A. fumigatus. Our results revealed that the β2-integrin knockdown was restricted to PMN and that CD18Ly6G cKO mice showed an aggravated course of IPA. In accordance, we observed a higher fungal burden and lower levels of proinflammatory innate cytokines, such as TNF-α, in lungs of IPA-infected CD18Ly6G cKO mice. Bronchoalveolar lavage revealed higher levels of CXCL1, a stronger PMN-infiltration, but concomitantly elevated apoptosis of PMN in lungs of CD18Ly6G cKO mice. Ex-vivo analysis further unveiled a strong impairment of PMN effector function, as reflected by an attenuated phagocytic activity, and a diminished generation of reactive oxygen species (ROS) and neutrophil-extracellular traps (NET) in CD18-deficient PMN. Overall, our study demonstrates that β2-integrins are required specifically for PMN effector functions and contribute to the clearance of A. fumigatus by infiltrating PMN, and the establishment of an inflammatory microenvironment in infected lungs.
Collapse
Affiliation(s)
- Maximilian Haist
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- *Correspondence: Maximilian Haist,
| | - Frederic Ries
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen, Germany
- Leibniz-Institut für Analytische Wissenschaften ISAS -e.V, Dortmund, Germany
| | - Monika Bednarczyk
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Ekkehard Siegel
- Institute for Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Michael Kuske
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Markus Radsak
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Daniel Teschner
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
27
|
Nording H, Sauter M, Lin C, Steubing R, Geisler S, Sun Y, Niethammer J, Emschermann F, Wang Y, Zieger B, Nieswandt B, Kleinschnitz C, Simon DI, Langer HF. Activated Platelets Upregulate β 2 Integrin Mac-1 (CD11b/CD18) on Dendritic Cells, Which Mediates Heterotypic Cell-Cell Interaction. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1729-1741. [PMID: 35277420 DOI: 10.4049/jimmunol.2100557] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 01/11/2022] [Indexed: 12/30/2022]
Abstract
Recent evidence suggests interaction of platelets with dendritic cells (DCs), while the molecular mechanisms mediating this heterotypic cell cross-talk are largely unknown. We evaluated the role of integrin Mac-1 (αMβ2, CD11b/CD18) on DCs as a counterreceptor for platelet glycoprotein (GP) Ibα. In a dynamic coincubation model, we observed interaction of human platelets with monocyte-derived DCs, but also that platelet activation induced a sharp increase in heterotypic cell binding. Inhibition of CD11b or GPIbα led to significant reduction of DC adhesion to platelets in vitro independent of GPIIbIIIa, which we confirmed using platelets from Glanzmann thrombasthenia patients and transgenic mouse lines on C57BL/6 background (GPIbα-/-, IL4R-GPIbα-tg, and muMac1 mice). In vivo, inhibition or genetic deletion of CD11b and GPIbα induced a significant reduction of platelet-mediated DC adhesion to the injured arterial wall. Interestingly, only intravascular antiCD11b inhibited DC recruitment, suggesting a dynamic DC-platelet interaction. Indeed, we could show that activated platelets induced CD11b upregulation on Mg2+-preactivated DCs, which was related to protein kinase B (Akt) and dependent on P-selectin and P-selectin glycoprotein ligand 1. Importantly, specific pharmacological targeting of the GPIbα-Mac-1 interaction site blocked DC-platelet interaction in vitro and in vivo. These results demonstrate that cross-talk of platelets with DCs is mediated by GPIbα and Mac-1, which is upregulated on DCs by activated platelets in a P-selectin glycoprotein ligand 1-dependent manner.
Collapse
Affiliation(s)
- Henry Nording
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany.,German Research Centre for Cardiovascular Research, Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany.,University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Manuela Sauter
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany.,University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Chaolan Lin
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Rebecca Steubing
- Department of Neurology and Center for Translational and Behavioral Neurosciences, University Hospital Essen, Essen, Germany
| | - Sven Geisler
- Cell Analysis Core Facility, University of Lübeck, Lübeck, Germany
| | - Ying Sun
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Joel Niethammer
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Fréderic Emschermann
- Department of Cardiovascular Medicine, University Hospital, Eberhard Karls University, Tübingen, Germany
| | - Yunmei Wang
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine and Harrington Heart & Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Barbara Zieger
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; and
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational and Behavioral Neurosciences, University Hospital Essen, Essen, Germany
| | - Daniel I Simon
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine and Harrington Heart & Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH.,University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Harald F Langer
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany; .,German Research Centre for Cardiovascular Research, Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany.,University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| |
Collapse
|
28
|
Peruń A, Gębicka M, Biedroń R, Skalska P, Józefowski S. The CD36 and SR-A/CD204 scavenger receptors fine-tune Staphylococcus aureus-stimulated cytokine production in mouse macrophages. Cell Immunol 2022; 372:104483. [DOI: 10.1016/j.cellimm.2022.104483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/20/2021] [Accepted: 01/11/2022] [Indexed: 11/03/2022]
|
29
|
Fu Y, Wang Z, Yu B, Lin Y, Huang E, Liu R, Zhao C, Lu M, Xu W, Liu H, Liu Y, Wang L, Chu Y. Intestinal CD11b + B Cells Ameliorate Colitis by Secreting Immunoglobulin A. Front Immunol 2021; 12:697725. [PMID: 34804004 PMCID: PMC8595478 DOI: 10.3389/fimmu.2021.697725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
The intestinal mucosal immune environment requires multiple immune cells to maintain homeostasis. Although intestinal B cells are among the most important immune cells, little is known about the mechanism that they employ to regulate immune homeostasis. In this study, we found that CD11b+ B cells significantly accumulated in the gut lamina propria and Peyer's patches in dextran sulfate sodium-induced colitis mouse models and patients with ulcerative colitis. Adoptive transfer of CD11b+ B cells, but not CD11b-/- B cells, effectively ameliorated colitis and exhibited therapeutic effects. Furthermore, CD11b+ B cells were found to produce higher levels of IgA than CD11b- B cells. CD11b deficiency in B cells dampened IgA production, resulting in the loss of their ability to ameliorate colitis. Mechanistically, CD11b+ B cells expressed abundant TGF-β and TGF-β receptor II, as well as highly activate phosphorylated Smad2/3 signaling pathway, consequently promoting the class switch to IgA. Collectively, our findings demonstrate that CD11b+ B cells are essential intestinal suppressive immune cells and the primary source of intestinal IgA, which plays an indispensable role in maintaining intestinal homeostasis.
Collapse
Affiliation(s)
- Ying Fu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhiming Wang
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Baichao Yu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yuli Lin
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Enyu Huang
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Pathology, The University of Hong Kong, Hong Kong, Hong Kong, SAR China
| | - Ronghua Liu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chujun Zhao
- Department of Biomedical Engineering, Columbia University, New York, NY, United States
| | - Mingfang Lu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wei Xu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hongchun Liu
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yongzhong Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Luman Wang
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Department of Endocrinology and Metabolism, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.,Biotherapy Research Center, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Xia P, Wu Y, Lian S, Yan L, Meng X, Duan Q, Zhu G. Research progress on Toll-like receptor signal transduction and its roles in antimicrobial immune responses. Appl Microbiol Biotechnol 2021; 105:5341-5355. [PMID: 34180006 PMCID: PMC8236385 DOI: 10.1007/s00253-021-11406-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/07/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022]
Abstract
When microorganisms invade a host, the innate immune system first recognizes the pathogen-associated molecular patterns of these microorganisms through pattern recognition receptors (PRRs). Toll-like receptors (TLRs) are known transmembrane PRRs existing in both invertebrates and vertebrates. Upon ligand recognition, TLRs initiate a cascade of signaling events; promote the pro-inflammatory cytokine, type I interferon, and chemokine expression; and play an essential role in the modulation of the host's innate and adaptive immunity. Therefore, it is of great significance to improve our understanding of antimicrobial immune responses by studying the role of TLRs and their signal molecules in the host's defense against invading microbes. This paper aims to summarize the specificity of TLRs in recognition of conserved microbial components, such as lipoprotein, lipopolysaccharide, flagella, endosomal nucleic acids, and other bioactive metabolites derived from microbes. This set of interactions helps to elucidate the immunomodulatory effect of TLRs and the signal transduction changes involved in the infectious process and provide a novel therapeutic strategy to combat microbial infections.
Collapse
Affiliation(s)
- Pengpeng Xia
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Yunping Wu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Siqi Lian
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Li Yan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Xia Meng
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Qiangde Duan
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| | - Guoqiang Zhu
- College of Veterinary Medicine (Institute of Comparative Medicine), Yangzhou University, 12th East Wenhui Road, Yangzhou, 225009 China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009 China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, 225009 China
| |
Collapse
|
31
|
Schittenhelm L, Robertson J, Pratt AG, Hilkens CM, Morrison VL. Dendritic cell integrin expression patterns regulate inflammation in the rheumatoid arthritis joint. Rheumatology (Oxford) 2021; 60:1533-1542. [PMID: 33123735 PMCID: PMC7937020 DOI: 10.1093/rheumatology/keaa686] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 09/19/2020] [Indexed: 11/14/2022] Open
Abstract
Objectives Immune dysregulation contributes to the development of RA. Altered surface expression patterns of integrin adhesion receptors by immune cells is one mechanism by which this may occur. We investigated the role of β2 integrin subunits CD11a and CD11b in dendritic cell (DC) subsets of RA patients. Methods Total β2 integrin subunit expression and its conformation (‘active’ vs ‘inactive’ state) were quantified in DC subsets from peripheral blood (PB) and SF of RA patients as well as PB from healthy controls. Ex vivo stimulation of PB DC subsets and in vitro-generated mature and tolerogenic monocyte-derived DCs (moDCs) were utilized to model the clinical findings. Integrin subunit contribution to DC function was tested by analysing clustering and adhesion, and in co-cultures to assess T cell activation. Results A significant reduction in total and active CD11a expression in DCs in RA SF compared with PB and, conversely, a significant increase in CD11b expression was found. These findings were modelled in vitro using moDCs: tolerogenic moDCs showed higher expression of active CD11a and reduced levels of active CD11b compared with mature moDCs. Finally, blockade of CD11b impaired T cell activation in DC–T cell co-cultures. Conclusion For the first time in RA, we show opposing expression of CD11a and CD11b in DCs in environments of inflammation (CD11alow/CD11bhigh) and steady state/tolerance (CD11ahigh/CD11blow), as well as a T cell stimulatory role for CD11b. These findings highlight DC integrins as potential novel targets for intervention in RA.
Collapse
Affiliation(s)
- Leonie Schittenhelm
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK.,Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK
| | - Jamie Robertson
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Arthur G Pratt
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK.,Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Catharien M Hilkens
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK
| | - Vicky L Morrison
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK
| |
Collapse
|
32
|
Wang T, Wang L, Zhu X, Liu G. Correlation between TLR4 gene polymorphism and acute respiratory distress syndrome after esophageal cancer surgery. Am J Transl Res 2021; 13:3337-3343. [PMID: 34017507 PMCID: PMC8129334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/10/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND One of the major postoperative complications of esophageal cancer is acute respiratory distress syndrome (ARDS), which poses a great threat to patients' lives. In this research, the cause of ARDS after esophageal cancer surgery was explained from the aspect of the single-nucleotide polymorphism at rs7873784, rs10759930 and rs10983755 of the Toll-like receptor 4 (TLR4) gene. METHODS A total of 75 patients complicated with ARDS after esophageal cancer surgery in our hospital were collected as the ARDS group and 150 patients without ARDS after surgery as the control group. Deoxyribonucleic acids (DNAs) in the peripheral blood of patients were extracted, and the polymorphism loci (rs7873784, rs10759930 and rs10983755) of the TLR4 gene were amplified through polymerase chain reaction (PCR) and sent to a company for sequencing. The concentration of serum TLR4 was detected by kits. RESULTS The frequencies of the G allele at rs7873784 (P=0.011) and C allele at rs10759930 (P=0.000) in the ARDS group were remarkably lower than those in the control group. Besides, the frequencies of GG genotype at rs7873784 (P=0.000) and CC and CT genotypes at rs10759930 (P=0.000) in the control group were notably higher than those in the ARDS group, while the frequency of AA genotype at rs10983755 (P=0.001) in the ARDS group was clearly lower than that in control group. The survival status of patients with complications of ARDS was notably correlated with CT genotype at rs10759930 of the TLR4 gene since patients with genotype CT were more likely to die (P=0.001). The GG genotype at rs10983755 of the TLR4 gene was remarkably related to the mean mechanical ventilation time (P=0.003) and the average length of intensive care unit (ICU) stay (P=0.018). The ARDS group had a lower frequency of GCG haplotype (P=0.009) and a higher frequency of GTA haplotype (P=0.001) than the control group. The linkage disequilibrium D' was 0.781 between rs7873784 and rs10759930 of the TLR4 gene, and two loci were linked to each other. In addition, the concentration of serum TRL4 in patients with genotype CC at rs7873784 (P=0.034), genotype CT at rs7873784 (P=0.000) and genotype GG at rs10983755 (P=0.000) of the TLR4 gene in the ARDS group was higher than that in the control group. CONCLUSION The single-nucleotide polymorphisms at rs7873784, rs10759930 and rs10983755 of the TLR4 gene are significantly related to ARDS after esophageal cancer surgery.
Collapse
Affiliation(s)
- Tongxin Wang
- Department of Emergency, People’s Hospital of Linyi Economic and Technological Development ZoneLinyi, Shandong Province, China
| | - Lijun Wang
- Department of Intensive Care Unit, People’s Hospital of Linyi Economic and Technological Development ZoneLinyi, Shandong Province, China
| | - Xiaojie Zhu
- Department of Internal Medicine, Shandong Coal Linyi Hot Spring SanatoriumLinyi, Shandong Province, China
| | - Gengxi Liu
- Department of Neurology, People’s Hospital of Linyi Economic and Technological Development ZoneLinyi, Shandong Province, China
| |
Collapse
|
33
|
Eslami-Kaliji F, Sarafbidabad M, Kiani-Esfahani A, Mirahmadi-Zare SZ, Dormiani K. 10-hydroxy-2-decenoic acid a bio-immunomodulator in tissue engineering; generates tolerogenic dendritic cells by blocking the toll-like receptor4. J Biomed Mater Res A 2021; 109:1575-1587. [PMID: 33638611 DOI: 10.1002/jbm.a.37152] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/03/2021] [Accepted: 02/10/2021] [Indexed: 01/22/2023]
Abstract
Dendritic cells (DCs), in response to the biomaterials, utilize toll-like receptors (TLRs) to become mature or tolerogenic through TLRs-dependent signaling pathways, especially TLR4. Regarding the physicochemical properties of biomaterials, some of such signaling pathways are activated. Unsaturated fatty acids have been explored as an antagonist for TLRs and lead to the tolerogenic phenotype of DCs. Here we showed that, although cultured DCs on both chitosan and Alginate-polyethyleneimine (Alg-PEI) films became fully mature, 10-hydroxy-2-decanoic acid (10-HDA), an unsaturated fatty acid found in royal jelly, led to the tolerogenic immunophenotype of DCs on both films. The cultured cells on the films possessed iDCs-like morphology in the presence of 10-HDA. Moreover, 10-HDA expressed lower levels of CD80, CD83, CD86, and HLA-DR, a higher level of IL-10, and lower level of IL-12 in the cultured DCs on both films. Furthermore, HEK293T cells expressing only TLR4 (HEK-TLR4 cells) were co-cultured with LPS, a specific agonist for TLR4, and 10-HDA. The 10-HDA significantly reduced the expression of tumor necrosis factor-a (TNF-α) in the HEK-TLR4 cells compared to treated only with LPS. These findings indicate that the 10-HDA acts as an antagonist of TLR4; therefore, potentially can be used in autoimmune diseases and preventing the rejection of biomaterials implantation and allograft transplantation.
Collapse
Affiliation(s)
- Farshid Eslami-Kaliji
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan, Iran.,Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohsen Sarafbidabad
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan, Iran
| | - Abbas Kiani-Esfahani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Seyede Zohreh Mirahmadi-Zare
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Kianoush Dormiani
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
34
|
Sim H, Jeong D, Kim HI, Pak S, Thapa B, Kwon HJ, Lee K. CD11b Deficiency Exacerbates Methicillin-Resistant Staphylococcus aureus-Induced Sepsis by Upregulating Inflammatory Responses of Macrophages. Immune Netw 2021; 21:e13. [PMID: 33996169 PMCID: PMC8099615 DOI: 10.4110/in.2021.21.e13] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/21/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
Macrophages are important for the first line of defense against microbial pathogens. Integrin CD11b, which is encoded by Itgam, is expressed on the surface of macrophages and has been implicated in adhesion, migration, and cell-mediated cytotoxicity. However, the functional impact of CD11b on the inflammatory responses of macrophages upon microbial infection remains unclear. Here, we show that CD11b deficiency resulted in increased susceptibility to sepsis induced by methicillin-resistant Staphylococcus aureus (MRSA) infection by enhancing the pro-inflammatory activities of macrophages. Upon infection with MRSA, the mortality of Itgam knockout mice was significantly higher than that of control mice, which is associated with increased production of TNF-α and IL-6. In response to MRSA, both bone marrow-derived macrophages and peritoneal macrophages lacking CD11b produced elevated amounts of pro-inflammatory cytokines and nitric oxide. Moreover, CD11b deficiency upregulated IL-4-induced expression of anti-inflammatory mediators such as IL-10 and arginase-1, and an immunomodulatory function of macrophages to restrain T cell activation. Biochemical and confocal microscopy data revealed that CD11b deficiency augmented the activation of NF-κB signaling and phosphorylation of Akt, which promotes the functional activation of macrophages with pro-inflammatory and immunoregulatory phenotypes, respectively. Overall, our experimental evidence suggests that CD11b is a critical modulator of macrophages in response to microbial infection.
Collapse
Affiliation(s)
- Hyunsub Sim
- Department of Biomedical Science, College of Natural Science, Hallym University, Chuncheon 24252, Korea
| | - Daecheol Jeong
- Department of Biomedical Science, College of Natural Science, Hallym University, Chuncheon 24252, Korea
| | - Hye-In Kim
- Department of Biomedical Science, College of Natural Science, Hallym University, Chuncheon 24252, Korea
| | - Seongwon Pak
- Department of Biomedical Science, College of Natural Science, Hallym University, Chuncheon 24252, Korea
| | - Bikash Thapa
- Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyung-Joo Kwon
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Keunwook Lee
- Department of Biomedical Science, College of Natural Science, Hallym University, Chuncheon 24252, Korea.,Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
35
|
Frautschy SA. CR3 ruffles FcγR's claim over phagocytic cups. J Biol Chem 2021; 296:100801. [PMID: 34019878 PMCID: PMC8191298 DOI: 10.1016/j.jbc.2021.100801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Phagocytosis plays diverse roles in biology, but our understanding of the purpose, interplay, and cell signaling mechanisms associated with different modes of phagocytosis is limited, without being able to capture and visualize each step in this rapid process from the beginning to end. A new study by Walbaum et al. uses stunning time-lapse 3D imaging of the engulfment of erythrocytes by macrophages via sinking, ruffling, and cup formation, unequivocally confirming a visionary 44-year-old theory derived from still electron microscopy photos that phagocytosis mediated by complement receptor CR3 occurs via a sinking mechanism and antibody-mediated phagocytosis occurs via phagocytic cup formation. The article also challenges the dogma, showing that phagocytic cup formation is not unique to antibody receptor phagocytosis, rather CR3 plays a complex role in different modes of phagocytosis. For example, inhibition of antibody-mediated phagocytosis leads to a compensatory upregulation of CR3-mediated sinking phagocytosis. These findings animate, in vivid colors, processes previously only captured as stills, exposing interactions between different phagocytic mechanisms and altering our basic understanding of this important process.
Collapse
Affiliation(s)
- S A Frautschy
- Geriatric Research Education and Clinical Center, Veterans Greater Los Angeles HealthCare System, Los Angeles, California, USA; Departments of Neurology and Medicine, University of California, Los Angeles, Los Angeles, California, USA.
| |
Collapse
|
36
|
|
37
|
Regulatory role of Gpr84 in the switch of alveolar macrophages from CD11b lo to CD11b hi status during lung injury process. Mucosal Immunol 2020; 13:892-907. [PMID: 32719411 DOI: 10.1038/s41385-020-0321-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 02/04/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a kind of comprehensive disease with excessive inflammation and high clinical mortality. Multiple immune cells are involved in the ARDS process. Amongst these populations, lung-resident alveolar macrophages (AMs) are known to participate in the regulation of ARDS. GPR84, a metabolite-sensing GPCR sensing medium-chain fatty acids (MCFAs), is highly expressed in LPS-challenged macrophages and considered as a pro-inflammatory receptor. In this study, it was hypothesized that Gpr84 may be involved in pulmonary homeostasis via its regulatory effect on the switch of AM status. In LPS-induced ALI mouse model, we identified the internal LPS-induced switch of AMs from CD11blo to more inflamed CD11bhi status, which is deeply related to the exacerbated imbalance of homeostasis in the lung injury process. Gpr84 was highly expressed in ALI lung tissues and involved in cytokine release, phagocytosis and status switch of AMs through positive regulatory crosstalk with TLR4-related pathways via CD14 and LBP, which relied on Akt, Erk1/2, and STAT3. If conserved in humans, GPR84 may represent a potential therapeutic target for ARDS.
Collapse
|
38
|
Ciesielska A, Matyjek M, Kwiatkowska K. TLR4 and CD14 trafficking and its influence on LPS-induced pro-inflammatory signaling. Cell Mol Life Sci 2020; 78:1233-1261. [PMID: 33057840 PMCID: PMC7904555 DOI: 10.1007/s00018-020-03656-y] [Citation(s) in RCA: 685] [Impact Index Per Article: 137.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/25/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Toll-like receptor (TLR) 4 belongs to the TLR family of receptors inducing pro-inflammatory responses to invading pathogens. TLR4 is activated by lipopolysaccharide (LPS, endotoxin) of Gram-negative bacteria and sequentially triggers two signaling cascades: the first one involving TIRAP and MyD88 adaptor proteins is induced in the plasma membrane, whereas the second engaging adaptor proteins TRAM and TRIF begins in early endosomes after endocytosis of the receptor. The LPS-induced internalization of TLR4 and hence also the activation of the TRIF-dependent pathway is governed by a GPI-anchored protein, CD14. The endocytosis of TLR4 terminates the MyD88-dependent signaling, while the following endosome maturation and lysosomal degradation of TLR4 determine the duration and magnitude of the TRIF-dependent one. Alternatively, TLR4 may return to the plasma membrane, which process is still poorly understood. Therefore, the course of the LPS-induced pro-inflammatory responses depends strictly on the rates of TLR4 endocytosis and trafficking through the endo-lysosomal compartment. Notably, prolonged activation of TLR4 is linked with several hereditary human diseases, neurodegeneration and also with autoimmune diseases and cancer. Recent studies have provided ample data on the role of diverse proteins regulating the functions of early, late, and recycling endosomes in the TLR4-induced inflammation caused by LPS or phagocytosis of E. coli. In this review, we focus on the mechanisms of the internalization and intracellular trafficking of TLR4 and CD14, and also of LPS, in immune cells and discuss how dysregulation of the endo-lysosomal compartment contributes to the development of diverse human diseases.
Collapse
Affiliation(s)
- Anna Ciesielska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland.
| | - Marta Matyjek
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| |
Collapse
|
39
|
Lim TJF, Bunjamin M, Ruedl C, Su IH. Talin1 controls dendritic cell activation by regulating TLR complex assembly and signaling. J Exp Med 2020; 217:e20191810. [PMID: 32438408 PMCID: PMC7398162 DOI: 10.1084/jem.20191810] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/24/2020] [Accepted: 03/27/2020] [Indexed: 12/29/2022] Open
Abstract
Talin critically controls integrin-dependent cell migration, but its regulatory role in skin dendritic cells (DCs) during inflammatory responses has not been investigated. Here, we show that talin1 regulates not only integrin-dependent Langerhans cell (LC) migration, but also MyD88-dependent Toll-like receptor (TLR)-stimulated DC activation. Talin1-deficient LCs failed to exit the epidermis, resulting in reduced LC migration to skin-draining lymph nodes (sdLNs) and defective skin tolerance induction, while talin1-deficient dermal DCs unexpectedly accumulated in the dermis despite their actomyosin-dependent migratory capabilities. Furthermore, talin1-deficient DCs exhibited compromised chemotaxis, NFκB activation, and proinflammatory cytokine production. Mechanistically, talin1 was required for the formation of preassembled TLR complexes in DCs at steady state via direct interaction with MyD88 and PIP5K. Local production of PIP2 by PIP5K then recruited TIRAP to the preassembled complexes, which were required for TLR signalosome assembly during DC activation. Thus, talin1 regulates MyD88-dependent TLR signaling pathways in DCs through a novel mechanism with implications for antimicrobial and inflammatory immune responses.
Collapse
Affiliation(s)
- Thomas Jun Feng Lim
- Laboratory of Molecular Immunology & Cell Signalling, School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Republic of Singapore
| | - Maegan Bunjamin
- Laboratory of Molecular Immunology & Cell Signalling, School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Republic of Singapore
| | - Christiane Ruedl
- Laboratory of Immunology, School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Republic of Singapore
| | - I-hsin Su
- Laboratory of Molecular Immunology & Cell Signalling, School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Republic of Singapore
| |
Collapse
|
40
|
Liu D, Ford ML. CD11b is a novel alternate receptor for CD154 during alloimmunity. Am J Transplant 2020; 20:2216-2225. [PMID: 32149455 PMCID: PMC7395865 DOI: 10.1111/ajt.15835] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/14/2020] [Accepted: 02/04/2020] [Indexed: 01/25/2023]
Abstract
Antagonism of the CD154/CD40 pathway is a highly effective means of inducing long-term graft survival in preclinical models. Using a fully allogeneic murine transplant model, we found that CD154 blockade was more effective in prolonging graft survival than was CD40 blockade, raising the possibility that CD154 binds a second receptor. To test this, we queried the impact of CD154 antagonism in the absence of CD40. Data indicated that anti-CD154 functioned to reduce graft-infiltrating CD8+ T cells in both WT and CD40-/- hosts. Because it has recently been reported that CD154 can ligate CD11b, we addressed the impact of blocking CD154-CD11b interactions during transplantation. We utilized a specific peptide antagonist that prevents CD154 binding of CD11b but has no effect on CD154-CD40 interactions. CD154:CD11b antagonism significantly increased the efficacy of anti-CD40 in prolonging allograft survival as compared to anti-CD40 plus control peptide. Mechanistically, CD154:CD11b antagonism functioned to reduce the frequency of graft-infiltrating CD8+ T cells and innate immune cells. These data therefore demonstrate that blocking CD154 interactions with both CD40 and CD11b is required for optimal inhibition of alloimmunity and provide an explanation for why CD40 blockers may be less efficacious than anti-CD154 reagents for the inhibition of allograft rejection.
Collapse
Affiliation(s)
- Danya Liu
- Emory Transplant Center and Department of Surgery, Emory University, Atlanta, Georgia
| | - Mandy L Ford
- Emory Transplant Center and Department of Surgery, Emory University, Atlanta, Georgia
| |
Collapse
|
41
|
Soto JA, Gálvez NMS, Andrade CA, Pacheco GA, Bohmwald K, Berrios RV, Bueno SM, Kalergis AM. The Role of Dendritic Cells During Infections Caused by Highly Prevalent Viruses. Front Immunol 2020; 11:1513. [PMID: 32765522 PMCID: PMC7378533 DOI: 10.3389/fimmu.2020.01513] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are a type of innate immune cells with major relevance in the establishment of an adaptive response, as they are responsible for the activation of lymphocytes. Since their discovery, several reports of their role during infectious diseases have been performed, highlighting their functions and their mechanisms of action. DCs can be categorized into different subsets, and each of these subsets expresses a wide arrange of receptors and molecules that aid them in the clearance of invading pathogens. Interferon (IFN) is a cytokine -a molecule of protein origin- strongly associated with antiviral immune responses. This cytokine is secreted by different cell types and is fundamental in the modulation of both innate and adaptive immune responses against viral infections. Particularly, DCs are one of the most important immune cells that produce IFN, with type I IFNs (α and β) highlighting as the most important, as they are associated with viral clearance. Type I IFN secretion can be induced via different pathways, activated by various components of the virus, such as surface proteins or genetic material. These molecules can trigger the activation of the IFN pathway trough surface receptors, including IFNAR, TLR4, or some intracellular receptors, such as TLR7, TLR9, and TLR3. Here, we discuss various types of dendritic cells found in humans and mice; their contribution to the activation of the antiviral response triggered by the secretion of IFN, through different routes of the induction for this important antiviral cytokine; and as to how DCs are involved in human infections that are considered highly frequent nowadays.
Collapse
Affiliation(s)
- Jorge A Soto
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolas M S Gálvez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A Andrade
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gaspar A Pacheco
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karen Bohmwald
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roslye V Berrios
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Instituto Milenio de Inmunología e Inmunoterapia, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
42
|
Li Y, Zhai P, Zheng Y, Zhang J, Kellum JA, Peng Z. Csf2 Attenuated Sepsis-Induced Acute Kidney Injury by Promoting Alternative Macrophage Transition. Front Immunol 2020; 11:1415. [PMID: 32733471 PMCID: PMC7358306 DOI: 10.3389/fimmu.2020.01415] [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] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a systemic inflammatory state that occurs in response to infection and significantly increases mortality in combination with acute kidney injury (AKI). Macrophages accumulate in the kidney after injury and undergo a transition from a proinflammatory (M1) phenotype to an alternatively activated (M2) phenotype that is required for normal repair. However, the specific signals that regulate the transition from the M1 to M2 phenotype in vivo are unknown. Here, we found an unexpected role of Colony stimulating factor 2 (Csf2) in controlling macrophage transition in vitro and in a mouse model of sepsis induced by cecal ligation and puncture (CLP). We first co-cultured human M1 macrophages with HK-2 cells and characterized cytokine/chemokine profiles via Luminex. Of the cytokines and chemokines that were overexpressed in medium from M1 macrophages cocultured with human kidney-2 (HK-2) cells compared with that from M1 macrophages cultured alone, Csf2 and IL6 showed the greatest increases. Csf2 was exclusively secreted by HK-2 cells but not by M1 macrophages. Furthermore, recombinant human Csf2 protein promoted transition of M1 macrophages to the M2 phenotype in a dose and time-dependent manner. The apoptosis and reactive oxygen species (ROS) release induced by M1 macrophages in HK-2 cells was attenuated after exposure to exogenous Csf2. In addition, the switch from the proinflammatory M1 phenotype to the M2 phenotype occurred via the p-Stat5 pathway, which was activated by Csf2. Importantly, we found that intraperitoneal injection of a Csf2-neutralizing antibody after CLP aggravated kidney injury and suppressed tubular proliferation, subsequently decreasing survival. However, administration of recombinant mouse Csf2 protein could rescue mice with sepsis. Together, our results indicate that Csf2 plays critical roles in regulating macrophage transition via activation of p-STAT5. These data form a foundation upon which new therapeutic strategies can be designed to improve the therapeutic efficacy of cytokine-based treatments for sepsis-induced AKI.
Collapse
Affiliation(s)
- Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Pan Zhai
- Department of Neurology, Hubei Province Hospital of Tradition Chinese Medicine, Wuhan, China
| | - Yawen Zheng
- Department of Urological Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jing Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - John A Kellum
- Center of Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Zhiyong Peng
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.,Center of Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
43
|
Mechanistic Understanding of Cell Recognition and Immune Reaction via CR1/CR3 by HAP- and SiO 2-NPs. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7474807. [PMID: 32382571 PMCID: PMC7195653 DOI: 10.1155/2020/7474807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 09/25/2019] [Accepted: 09/30/2019] [Indexed: 01/02/2023]
Abstract
Nanodrug carrier will eventually enter the blood when intravenously injected or in other ways. Meanwhile, a series of toxic effects were caused to the body with the formation of nanoparticle protein corona. In our studies, we try to reveal the recognition mechanism of nanoparticle protein corona by monocyte and the damage effect on immune cells by activated complement of hydroxyapatite nanoparticles (HAP-NPs) and silicon dioxide nanoparticles (SiO2-NPs). So expressions of TLR4/CR1/CR were analyzed by flow cytometry (FCM) in order to illuminate the recognition mechanism of nanoparticle protein corona by monocyte. And the expression of ROS, cytokines, adhesion molecules, and arachidonic acid was measured when THP-1 and HUVECs were stimulated by NP-activated complement. The results showed that HAP-NPs can be recognized by the opsonin receptor (iC3b/CR3) model, while plasma protein, opsonin receptor, and Toll-like receptors are all likely launch cell recognition of SiO2-NPs. And it was considerate that NP-activated complement can damage THP-1 and HUVECs, including oxidative stress, inflammation, and increased vascular permeability. So the surface of nanodrug carrier can be modified to avoid being clear and reduce the efficacy according to the three receptors (TLR4/CR1/CR3).
Collapse
|
44
|
Sin WX, Yeong JPS, Lim TJF, Su IH, Connolly JE, Chin KC. IRF-7 Mediates Type I IFN Responses in Endotoxin-Challenged Mice. Front Immunol 2020; 11:640. [PMID: 32373120 PMCID: PMC7176903 DOI: 10.3389/fimmu.2020.00640] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 03/20/2020] [Indexed: 01/23/2023] Open
Abstract
IRF-7 mediates robust production of type I IFN via MyD88 of the TLR9 pathway in plasmacytoid dendritic cells (pDCs). Previous in vitro studies using bone marrow-derived dendritic cells lacking either Irf7 or Irf3 have demonstrated that only IRF-3 is required for IFN-β production in the TLR4 pathway. Here, we show that IRF-7 is essential for both type I IFN induction and IL-1β responses via TLR4 in mice. Mice lacking Irf7 were defective in production of both IFN-β and IL-1β, an IFN-β-induced pro-inflammatory cytokine, after LPS challenge. IFN-β production in response to LPS was impaired in IRF-7-deficient macrophages, but not dendritic cells. Unlike pDCs, IRF-7 is activated by the TRIF-, but not MyD88-, dependent pathway via TBK-1 in macrophages after LPS stimulation. Like pDCs, resting macrophages constitutively expressed IRF-7 protein. This basal IRF-7 protein was completely abolished in either Ifnar1 -/- or Stat1 -/- macrophages, which corresponded with the loss of LPS-stimulated IFN-β induction in these macrophages. These findings demonstrate that macrophage IRF-7 is critical for LPS-induced type I IFN responses, which in turn facilitate IL-1β production in mice.
Collapse
Affiliation(s)
- Wei-Xiang Sin
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Joe Poh-Sheng Yeong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Thomas Jun Feng Lim
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Singapore
| | - I-Hsin Su
- School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Singapore
| | - John E Connolly
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,Institute of Biomedical Studies, Baylor University, Waco, TX, United States
| | - Keh-Chuang Chin
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Physiology, NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
45
|
Abstract
The recognition of microbial or danger-associated molecular patterns by complement proteins initiates a cascade of events that culminates in the activation of surface complement receptors on immune cells. Such signalling pathways converge with those activated downstream of pattern recognition receptors to determine the type and magnitude of the immune response. Intensive investigation in the field has uncovered novel pathways that link complement-mediated signalling with homeostatic and pathological T cell responses. More recently, the observation that complement proteins also act in the intracellular space to shape T cell fates has added a new layer of complexity. Here, we consider fundamental mechanisms and novel concepts at the interface of complement biology and immunity and discuss how these affect the maintenance of homeostasis and the development of human pathology.
Collapse
|
46
|
Bednarczyk M, Stege H, Grabbe S, Bros M. β2 Integrins-Multi-Functional Leukocyte Receptors in Health and Disease. Int J Mol Sci 2020; 21:E1402. [PMID: 32092981 PMCID: PMC7073085 DOI: 10.3390/ijms21041402] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 12/25/2022] Open
Abstract
β2 integrins are heterodimeric surface receptors composed of a variable α (CD11a-CD11d) and a constant β (CD18) subunit and are specifically expressed by leukocytes. The α subunit defines the individual functional properties of the corresponding β2 integrin, but all β2 integrins show functional overlap. They mediate adhesion to other cells and to components of the extracellular matrix (ECM), orchestrate uptake of extracellular material like complement-opsonized pathogens, control cytoskeletal organization, and modulate cell signaling. This review aims to delineate the tremendous role of β2 integrins for immune functions as exemplified by the phenotype of LAD-I (leukocyte adhesion deficiency 1) patients that suffer from strong recurrent infections. These immune defects have been largely attributed to impaired migratory and phagocytic properties of polymorphonuclear granulocytes. The molecular base for this inherited disease is a functional impairment of β2 integrins due to mutations within the CD18 gene. LAD-I patients are also predisposed for autoimmune diseases. In agreement, polymorphisms within the CD11b gene have been associated with autoimmunity. Consequently, β2 integrins have received growing interest as targets in the treatment of autoimmune diseases. Moreover, β2 integrin activity on leukocytes has been implicated in tumor development.
Collapse
Affiliation(s)
| | | | | | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (H.S.); (S.G.)
| |
Collapse
|
47
|
Integrin CD11b Deficiency Aggravates Retinal Microglial Activation and RGCs Degeneration After Acute Optic Nerve Injury. Neurochem Res 2020; 45:1072-1085. [PMID: 32052258 DOI: 10.1007/s11064-020-02984-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/23/2020] [Accepted: 02/08/2020] [Indexed: 12/13/2022]
Abstract
Neuroinflammation plays a vital role in the process of a variety of retinal ganglion cells (RGCs) degenerative diseases including traumatic optic neuropathy (TON). Retinal microglial activation is believed as a harbinger of TON, and robust microglial activation can aggravate trauma-induced RGCs degeneration, which ultimately leads to RGCs loss. Toll like receptor 4 (TLR4)-triggered inflammation is of great importance in retinal inflammatory response after optic nerve injury. CD11b on macrophage and brain microglia can inhibit TLR4-triggered inflammation. However, the functional role of CD11b in retinal microglia is not well understood. Here, using an optic nerve crush model and CD11b gene deficient mice, we found that CD11b protein expression was mainly on retinal microglia, significantly increased after optic nerve injury, and still maintained at a high level till at least 28 days post crush. Compared with wild type mice, following acute optic nerve injury, CD11b deficient retinae exhibited more exacerbated microglial activation, accelerated RGCs degeneration, less growth associated protein-43 expression, as well as more proinflammatory cytokines such as interleukin-6 and tumor necrosis factor α while less anti-inflammatory factors such as arginase-1 and interleukin-10 production. We conclude that CD11b is essential in regulating retinal microglial activation and neuroinflammatory responses after acute optic nerve injury, which is critical for subsequent RGCs degeneration and loss.
Collapse
|
48
|
Vilar R, Fish RJ, Casini A, Neerman-Arbez M. Fibrin(ogen) in human disease: both friend and foe. Haematologica 2020; 105:284-296. [PMID: 31949010 PMCID: PMC7012490 DOI: 10.3324/haematol.2019.236901] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/21/2019] [Indexed: 12/19/2022] Open
Abstract
Fibrinogen is an abundant protein synthesized in the liver, present in human blood plasma at concentrations ranging from 1.5-4 g/L in healthy individuals with a normal half-life of 3-5 days. With fibrin, produced by thrombin-mediated cleavage, fibrinogen plays important roles in many physiological processes. Indeed, the formation of a stable blood clot, containing polymerized and cross-linked fibrin, is crucial to prevent blood loss and drive wound healing upon vascular injury. A balance between clotting, notably the conversion of fibrinogen to fibrin, and fibrinolysis, the proteolytic degradation of the fibrin mesh, is essential. Disruption of this equilibrium can cause disease in distinct manners. While some pathological conditions are the consequence of altered levels of fibrinogen, others are related to structural properties of the molecule. The source of fibrinogen expression and the localization of fibrin(ogen) protein also have clinical implications. Low levels of fibrinogen expression have been detected in extra-hepatic tissues, including carcinomas, potentially contributing to disease. Fibrin(ogen) deposits at aberrant sites including the central nervous system or kidney, can also be pathological. In this review, we discuss disorders in which fibrinogen and fibrin are implicated, highlighting mechanisms that may contribute to disease.
Collapse
Affiliation(s)
- Rui Vilar
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine
| | - Richard J Fish
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine
| | - Alessandro Casini
- Division of Angiology and Hemostasis, University Hospitals and University of Geneva Faculty of Medicine
| | - Marguerite Neerman-Arbez
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine .,iGE3, Institute of Genetics and Genomics in Geneva, Geneva, Switzerland
| |
Collapse
|
49
|
Gruber EJ, Leifer CA. Molecular regulation of TLR signaling in health and disease: mechano-regulation of macrophages and TLR signaling. Innate Immun 2020; 26:15-25. [PMID: 31955624 PMCID: PMC6974875 DOI: 10.1177/1753425919838322] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/18/2019] [Accepted: 02/19/2019] [Indexed: 12/28/2022] Open
Abstract
Immune cells encounter tissues with vastly different biochemical and physical characteristics. Much of the research emphasis has focused on the role of cytokines and chemokines in regulating immune cell function, but the role of the physical microenvironment has received considerably less attention. The tissue mechanics, or stiffness, of healthy tissues varies dramatically from soft adipose tissue and brain to stiff cartilage and bone. Tissue mechanics also change due to fibrosis and with diseases such as atherosclerosis or cancer. The process by which cells sense and respond to their physical microenvironment is called mechanotransduction. Here we review mechanotransduction in immunologically important diseases and how physical characteristics of tissues regulate immune cell function, with a specific emphasis on mechanoregulation of macrophages and TLR signaling.
Collapse
Affiliation(s)
| | - Cynthia A Leifer
- Department of Microbiology and Immunology, Cornell
University, Ithaca, NY, USA
| |
Collapse
|
50
|
N Rosalez M, Estevez-Fregoso E, Alatorre A, Abad-García A, A Soriano-Ursúa M. 2-Aminoethyldiphenyl Borinate: A Multitarget Compound with Potential as a Drug Precursor. Curr Mol Pharmacol 2020; 13:57-75. [PMID: 31654521 DOI: 10.2174/1874467212666191025145429] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/30/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Boron is considered a trace element that induces various effects in systems of the human body. However, each boron-containing compound exerts different effects. OBJECTIVE To review the effects of 2-Aminoethyldiphenyl borinate (2-APB), an organoboron compound, on the human body, but also, its effects in animal models of human disease. METHODS In this review, the information to showcase the expansion of these reported effects through interactions with several ion channels and other receptors has been reported. These effects are relevant in the biomedical and chemical fields due to the application of the reported data in developing therapeutic tools to modulate the functions of the immune, cardiovascular, gastrointestinal and nervous systems. RESULTS Accordingly, 2-APB acts as a modulator of adaptive and innate immunity, including the production of cytokines and the migration of leukocytes. Additionally, reports show that 2-APB exerts effects on neurons, smooth muscle cells and cardiomyocytes, and it provides a cytoprotective effect by the modulation and attenuation of reactive oxygen species. CONCLUSION The molecular pharmacology of 2-APB supports both its potential to act as a drug and the desirable inclusion of its moieties in new drug development. Research evaluating its efficacy in treating pain and specific maladies, such as immune, cardiovascular, gastrointestinal and neurodegenerative disorders, is scarce but interesting.
Collapse
Affiliation(s)
- Melvin N Rosalez
- Department of Physiology, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Diaz Miron S/N, Mexico City, 11340, Mexico
| | - Elizabeth Estevez-Fregoso
- Department of Physiology, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Diaz Miron S/N, Mexico City, 11340, Mexico
| | - Alberto Alatorre
- Department of Physiology, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Diaz Miron S/N, Mexico City, 11340, Mexico
| | - Antonio Abad-García
- Department of Physiology, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Diaz Miron S/N, Mexico City, 11340, Mexico
| | - Marvin A Soriano-Ursúa
- Department of Physiology, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis and Diaz Miron S/N, Mexico City, 11340, Mexico
| |
Collapse
|