1
|
Baghaie L, Bunsick DA, Aucoin EB, Skapinker E, Yaish AM, Li Y, Harless WW, Szewczuk MR. Pro-Inflammatory Cytokines Transactivate Glycosylated Cytokine Receptors on Cancer Cells to Induce Epithelial-Mesenchymal Transition to the Metastatic Phenotype. Cancers (Basel) 2025; 17:1234. [PMID: 40227834 PMCID: PMC11988151 DOI: 10.3390/cancers17071234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025] Open
Abstract
Background/Objectives: The significance of cytokine signaling on cancer progression and metastasis has raised interest in cancer research over the last few decades. Here, we analyzed the effects of three cytokines that we previously reported are significantly upregulated rapidly after the surgical removal of primary breast, colorectal, and prostate cancer. We also investigated the regulation of their cognate receptors. Methods: All experiments were conducted using the PANC-1, SW620, and MCF-7 cell lines, treated with three different cytokines (TGF-β1, HGF, and IL-6). The effect of these cytokines on the expression of epithelial-mesenchymal transition (EMT) cell surface markers and neuraminidase-1 activity was measured via fluorescent microscopy and image analysis software. Results: The findings show that these cytokines increase the expression of mesenchymal markers while reducing epithelial markers, corresponding to the EMT process. A strong link between cytokine receptor signaling and the Neu-1-MMP-9-GPCR crosstalk was identified, suggesting that cytokine receptor binding leads to increased Neu-1 activity and subsequent signaling pathway activation. Oseltamivir phosphate (OP) prevented sialic acid hydrolysis by neuraminidase-1 (Neu-1), leading to the downregulation of these signaling cascades. Conclusions: In concert with the previous work revealing the role of Neu-1 in regulating other glycosylated receptors implicated in cancer cell proliferation and EMT, targeting Neu-1 may provide effective treatment against a variety of malignancies. Most significantly, the treatment of patients with specific inhibitors of Neu-1 soon after primary cancer surgery may improve our ability to cure early-stage cancer by inhibiting the EMT process and disrupting the ability of any residual cancer cell population to metastasize.
Collapse
Affiliation(s)
- Leili Baghaie
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (L.B.); (D.A.B.)
| | - David A. Bunsick
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (L.B.); (D.A.B.)
| | - Emilyn B. Aucoin
- Faculty of Science, Biology (Biomedical Science), York University, Toronto, ON M3J 1P3, Canada;
| | - Elizabeth Skapinker
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | | | - Yunfan Li
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | | | - Myron R. Szewczuk
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (L.B.); (D.A.B.)
| |
Collapse
|
2
|
Lialios P, Alimperti S. Role of E-cadherin in epithelial barrier dysfunction: implications for bacterial infection, inflammation, and disease pathogenesis. Front Cell Infect Microbiol 2025; 15:1506636. [PMID: 40007608 PMCID: PMC11850337 DOI: 10.3389/fcimb.2025.1506636] [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] [Received: 10/05/2024] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Epithelial barriers serve as critical defense lines against microbial infiltration and maintain tissue homeostasis. E-cadherin, an essential component of adherens junctions, has emerged as a pivotal molecule that secures epithelial homeostasis. Lately, its pleiotropic role beyond barrier function, including its involvement in immune responses, has become more evident. Herein, we delve into the intricate relationship between (dys)regulation of epithelial homeostasis and the versatile functionality of E-cadherin, describing complex mechanisms that underlie barrier integrity and disruption in disease pathogenesis such as bacterial infection and inflammation, among others. Clinical implications of E-cadherin perturbations in host pathophysiology are emphasized; downregulation, proteolytic phenomena, abnormal localization/signaling and aberrant immune reactions are linked with a broad spectrum of pathology beyond infectious diseases. Finally, potential therapeutic interventions that may harness E-cadherin to mitigate barrier-associated tissue damage are explored. Overall, this review highlights the crucial role of E-cadherin in systemic health, offering insights that could pave the way for strategies to reinforce/restore barrier integrity and treat related diseases.
Collapse
Affiliation(s)
- Peter Lialios
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
- Center for Biological and Biomedical Engineering, Georgetown University, Washington, DC, United States
| | - Stella Alimperti
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
- Center for Biological and Biomedical Engineering, Georgetown University, Washington, DC, United States
| |
Collapse
|
3
|
Tolomeo M, Cascio A. STAT4 and STAT6, their role in cellular and humoral immunity and in diverse human diseases. Int Rev Immunol 2024; 43:394-418. [PMID: 39188021 DOI: 10.1080/08830185.2024.2395274] [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: 06/29/2023] [Revised: 10/23/2023] [Accepted: 08/17/2024] [Indexed: 08/28/2024]
Abstract
Signal transducer and activator of transcription (STAT) 4 and STAT6 play a crucial role in immune cells by transducing signals from specific cytokine receptors, and inducing transcription of genes involved in cell-mediated and humoral immunity. These two different defense mechanisms against pathogens are regulated by two specific CD4+ T helper (Th) cells known as Th1 and Th2 cells. Many studies have shown that several diseases including cancer, inflammatory, autoimmune and allergic diseases are associated with a Th1/Th2 imbalance caused by increased or decreased expression/activity of STAT4 or STAT6 often due to genetic and epigenetic aberrances. An altered expression of STAT4 has been observed in different tumors and autoimmune diseases, while a dysregulation of STAT6 signaling pathway is frequently observed in allergic conditions, such as atopic dermatitis, allergic asthma, food allergy, and tumors such as Hodgkin and non-Hodgkin lymphomas. Recently, dysregulations of STAT4 and STAT6 expression have been observed in SARS-CoV2 and monkeypox infections, which are still public health emergencies in many countries. SARS-CoV-2 can induce an imbalance in Th1 and Th2 responses with a predominant activation of STAT6 in the cytosol and nuclei of pneumocytes that drives Th2 polarization and cytokine storm. In monkeypox infection the virus can promote an immune evasion by inducing a Th2 response that in turn inhibits the Th1 response essential for virus elimination. Furthermore, genetic variations of STAT4 that are associated with an increased risk of developing systemic lupus erythematosus seem to play a role in defense against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Manlio Tolomeo
- Department of Infectious Diseases, A.O.U.P. Palermo, Palermo, Italy
| | - Antonio Cascio
- Department of Infectious Diseases, A.O.U.P. Palermo, Palermo, Italy
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, Palermo, Italy
| |
Collapse
|
4
|
Leiba J, Sipka T, Begon-Pescia C, Bernardello M, Tairi S, Bossi L, Gonzalez AA, Mialhe X, Gualda EJ, Loza-Alvarez P, Blanc-Potard A, Lutfalla G, Nguyen-Chi ME. Dynamics of macrophage polarization support Salmonella persistence in a whole living organism. eLife 2024; 13:e89828. [PMID: 38224094 PMCID: PMC10830131 DOI: 10.7554/elife.89828] [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: 06/02/2023] [Accepted: 01/14/2024] [Indexed: 01/16/2024] Open
Abstract
Numerous intracellular bacterial pathogens interfere with macrophage function, including macrophage polarization, to establish a niche and persist. However, the spatiotemporal dynamics of macrophage polarization during infection within host remain to be investigated. Here, we implement a model of persistent Salmonella Typhimurium infection in zebrafish, which allows visualization of polarized macrophages and bacteria in real time at high resolution. While macrophages polarize toward M1-like phenotype to control early infection, during later stages, Salmonella persists inside non-inflammatory clustered macrophages. Transcriptomic profiling of macrophages showed a highly dynamic signature during infection characterized by a switch from pro-inflammatory to anti-inflammatory/pro-regenerative status and revealed a shift in adhesion program. In agreement with this specific adhesion signature, macrophage trajectory tracking identifies motionless macrophages as a permissive niche for persistent Salmonella. Our results demonstrate that zebrafish model provides a unique platform to explore, in a whole organism, the versatile nature of macrophage functional programs during bacterial acute and persistent infections.
Collapse
Affiliation(s)
- Jade Leiba
- LPHI, Université de Montpellier, CNRS, INSERMMontpellierFrance
| | - Tamara Sipka
- LPHI, Université de Montpellier, CNRS, INSERMMontpellierFrance
| | | | - Matteo Bernardello
- ICFO - Institute of Photonic Sciences, The Barcelona Institute of Science and TechnologyCastelldefels, BarcelonaSpain
| | - Sofiane Tairi
- LPHI, Université de Montpellier, CNRS, INSERMMontpellierFrance
| | - Lionello Bossi
- Institute for Integrative Biology of the Cell-I2BC, Université Paris-Saclay, CEA, CNRSGif-sur-YvetteFrance
| | - Anne-Alicia Gonzalez
- MGX-Montpellier GenomiX, Université de Montpellier, CNRS, INSERMMontpellierFrance
| | - Xavier Mialhe
- MGX-Montpellier GenomiX, Université de Montpellier, CNRS, INSERMMontpellierFrance
| | - Emilio J Gualda
- ICFO - Institute of Photonic Sciences, The Barcelona Institute of Science and TechnologyCastelldefels, BarcelonaSpain
| | - Pablo Loza-Alvarez
- ICFO - Institute of Photonic Sciences, The Barcelona Institute of Science and TechnologyCastelldefels, BarcelonaSpain
| | | | | | | |
Collapse
|
5
|
Lokwani R, Josyula A, Ngo TB, DeStefano S, Fertil D, Faust M, Adusei KM, Bhuiyan M, Lin A, Karkanitsa M, Maclean E, Fathi P, Su Y, Liu J, Vishwasrao HD, Sadtler K. Pro-regenerative biomaterials recruit immunoregulatory dendritic cells after traumatic injury. NATURE MATERIALS 2024; 23:147-157. [PMID: 37872423 DOI: 10.1038/s41563-023-01689-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 09/12/2023] [Indexed: 10/25/2023]
Abstract
During wound healing and surgical implantation, the body establishes a delicate balance between immune activation to fight off infection and clear debris and immune tolerance to control reactivity against self-tissue. Nonetheless, how such a balance is achieved is not well understood. Here we describe that pro-regenerative biomaterials for muscle injury treatment promote the proliferation of a BATF3-dependent CD103+XCR1+CD206+CD301b+ dendritic cell population associated with cross-presentation and self-tolerance. Upregulation of E-cadherin, the ligand for CD103, and XCL-1 in injured tissue suggests a mechanism for cell recruitment to trauma. Muscle injury recruited natural killer cells that produced Xcl1 when stimulated with fragmented extracellular matrix. Without cross-presenting cells, T-cell activation increases, pro-regenerative macrophage polarization decreases and there are alterations in myogenesis, adipogenesis, fibrosis and increased muscle calcification. These results, previously observed in cancer progression, suggest a fundamental mechanism of immune regulation in trauma and material implantation with implications for both short- and long-term injury recovery.
Collapse
Affiliation(s)
- Ravi Lokwani
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Aditya Josyula
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Tran B Ngo
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Sabrina DeStefano
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Daphna Fertil
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Mondreakest Faust
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth M Adusei
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Minhaj Bhuiyan
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Aaron Lin
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- Unit for Nanoengineering and Microphysiological Systems, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Maria Karkanitsa
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Efua Maclean
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Parinaz Fathi
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- Unit for Nanoengineering and Microphysiological Systems, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Yijun Su
- Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Jiamin Liu
- Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Harshad D Vishwasrao
- Advanced Imaging and Microscopy Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Kaitlyn Sadtler
- Section on Immunoengineering, Biomedical Engineering and Technology Acceleration Center, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
CD103 Expression on Regulatory and Follicular T Cells in Lymph Nodes, Bronchoalveolar Lavage Fluid and Peripheral Blood of Sarcoidosis Patients. Life (Basel) 2022; 12:life12050762. [PMID: 35629428 PMCID: PMC9146853 DOI: 10.3390/life12050762] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 11/26/2022] Open
Abstract
(1) Background: Sarcoidosis is a chronic multisystem disorder of unknown aetiology, driven by a T-cell mechanism allowing T-cell attachment and transmigration through the endothelium, and endorsed by the expression of an integrin alpha-E beta-7 (CD103). This study aimed to analyse the different distribution and compartmentalisation of CD103 expression on T cell subsets in BAL, peripheral blood mononuclear cells (PBMC) and lymph nodes (LLN) from sarcoidosis patients. (2) Patients: We consecutively and prospectively enrolled 14 sarcoidosis patients. We collected PBMC, LLN and BAL at the same time from all patients. Through flow cytometric analysis, we analysed the expression of CD103 on regulatory and follicular T cell subsets. (3) Results: All patients were in radiological Scadding stage II. The multivariate analysis found that the variables which most influenced the peripheral blood compartment were high CD8+ and low ThReg, CD8+CD103+ and Tfh cell percentages. A principal component analysis plot performed to distinguish LLN, BAL and PBMC showed that they separated on the basis of CD4+, CD4+CD103+, CD8+, CD8+CD103+, TcEffector, TcNaive, ThNaive, ThEffector, Threg, ThregCD103+, Tfh, TcfCXC5+ and CD4+CD103+/CD4+ with 65.96% of the total variance. (4) Conclusions: Our study is the first to report a link between the imbalance in circulating, alveolar and lymph node CD8+ and CD8+CD103+ T cells, ThReg, Tfh and ThNaive and the CD103+CD4+/CD4+ T cell ratio in the development of sarcoidosis. These findings shine a spotlight on the pathogenesis of sarcoidosis and may offer new predictors for diagnosis. Our study provides additional understanding for a personalised, and hopefully more effective treatment of sarcoidosis.
Collapse
|
7
|
Cronan MR. In the Thick of It: Formation of the Tuberculous Granuloma and Its Effects on Host and Therapeutic Responses. Front Immunol 2022; 13:820134. [PMID: 35320930 PMCID: PMC8934850 DOI: 10.3389/fimmu.2022.820134] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
The defining pathology of tuberculosis is the granuloma, an organized structure derived from host immune cells that surrounds infecting Mycobacterium tuberculosis. As the location of much of the bacteria in the infected host, the granuloma is a central point of interaction between the host and the infecting bacterium. This review describes the signals and cellular reprogramming that drive granuloma formation. Further, as a central point of host-bacterial interactions, the granuloma shapes disease outcome by altering host immune responses and bacterial susceptibility to antibiotic treatment, as discussed herein. This new understanding of granuloma biology and the signaling behind it highlights the potential for host-directed therapies targeting the granuloma to enhance antibiotic access and tuberculosis-specific immune responses.
Collapse
Affiliation(s)
- Mark R. Cronan
-
In Vivo Cell Biology of Infection Group, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
8
|
Hongo D, Zheng P, Dutt S, Pawar RD, Meyer E, Engleman EG, Strober S. Identification of Two Subsets of Murine DC1 Dendritic Cells That Differ by Surface Phenotype, Gene Expression, and Function. Front Immunol 2021; 12:746469. [PMID: 34777358 PMCID: PMC8589020 DOI: 10.3389/fimmu.2021.746469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/17/2021] [Indexed: 11/13/2022] Open
Abstract
Classical dendritic cells (cDCs) in mice have been divided into 2 major subsets based on the expression of nuclear transcription factors: a CD8+Irf8+Batf3 dependent (DC1) subset, and a CD8-Irf4+ (DC2) subset. We found that the CD8+DC1 subset can be further divided into CD8+DC1a and CD8+DC1b subsets by differences in surface receptors, gene expression, and function. Whereas all 3 DC subsets can act alone to induce potent Th1 cytokine responses to class I and II MHC restricted peptides derived from ovalbumin (OVA) by OT-I and OT-II transgenic T cells, only the DC1b subset could effectively present glycolipid antigens to natural killer T (NKT) cells. Vaccination with OVA protein pulsed DC1b and DC2 cells were more effective in reducing the growth of the B16-OVA melanoma as compared to pulsed DC1a cells in wild type mice. In conclusion, the Batf3-/- dependent DC1 cells can be further divided into two subsets with different immune functional profiles in vitro and in vivo.
Collapse
Affiliation(s)
- David Hongo
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
| | - Pingping Zheng
- Department of Medicine, Division of Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA, United States
| | - Suparna Dutt
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
| | - Rahul D Pawar
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
| | - Everett Meyer
- Department of Medicine, Division of Blood and Marrow Transplantation, Stanford University School of Medicine, Stanford, CA, United States
| | - Edgar G Engleman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Samuel Strober
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
9
|
Ahuja S, Lazar IM. Systems-Level Proteomics Evaluation of Microglia Response to Tumor-Supportive Anti-Inflammatory Cytokines. Front Immunol 2021; 12:646043. [PMID: 34566949 PMCID: PMC8458581 DOI: 10.3389/fimmu.2021.646043] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/06/2021] [Indexed: 12/24/2022] Open
Abstract
Background Microglia safeguard the CNS against injuries and pathogens, and in the presence of certain harmful stimuli are capable of inducing a disease-dependent inflammatory response. When exposed to anti-inflammatory cytokines, however, these cells possess the ability to switch from an inflammatory to an immunosuppressive phenotype. Cancer cells exploit this property to evade the immune system, and elicit an anti-inflammatory microenvironment that facilitates tumor attachment and growth. Objective The tumor-supportive biological processes that are activated in microglia cells in response to anti-inflammatory cytokines released from cancer cells were explored with mass spectrometry and proteomic technologies. Methods Serum-depleted and non-depleted human microglia cells (HMC3) were treated with a cocktail of IL-4, IL-13, IL-10, TGFβ, and CCL2. The cellular protein extracts were analyzed by LC-MS/MS. Using functional annotation clustering tools, statistically significant proteins that displayed a change in abundance between cytokine-treated and non-treated cells were mapped to their biological networks and pathways. Results The proteomic analysis of HMC3 cells enabled the identification of ~10,000 proteins. Stimulation with anti-inflammatory cytokines resulted in the activation of distinct, yet integrated clusters of proteins that trigger downstream a number of tumor-promoting biological processes. The observed changes could be classified into four major categories, i.e., mitochondrial gene expression, ECM remodeling, immune response, and impaired cell cycle progression. Intracellular immune activation was mediated mainly by the transducers of MAPK, STAT, TGFβ, NFKB, and integrin signaling pathways. Abundant collagen formation along with the expression of additional receptors, matrix components, growth factors, proteases and protease inhibitors, was indicative of ECM remodeling processes supportive of cell-cell and cell-matrix adhesion. Overexpression of integrins and their modulators was reflective of signaling processes that link ECM reorganization with cytoskeletal re-arrangements supportive of cell migration. Antigen processing/presentation was represented by HLA class I histocompatibility antigens, and correlated with upregulated proteasomal subunits, vesicular/viral transport, and secretory processes. Immunosuppressive and proangiogenic chemokines, as well as anti-angiogenic factors, were detectable in low abundance. Pronounced pro-inflammatory, chemotactic or phagocytic trends were not observed, however, the expression of certain receptors, signaling and ECM proteins indicated the presence of such capabilities. Conclusions Comprehensive proteomic profiling of HMC3 cells stimulated with anti-inflammatory cytokines revealed a spectrum of microglia phenotypes supportive of cancer development in the brain via microenvironment-dependent biological mechanisms.
Collapse
Affiliation(s)
- Shreya Ahuja
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
| | - Iulia M. Lazar
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, United States
- Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA, United States
- Carilion School of Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
10
|
Immunonutritional Protease Inhibitors from T. durum and A. sativa Display Metabolic Similarities When Assayed on Human Macrophage-like Cells. Int J Mol Sci 2021; 22:ijms22158307. [PMID: 34361073 PMCID: PMC8347451 DOI: 10.3390/ijms22158307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 12/20/2022] Open
Abstract
This study evaluated the immunonutritional effects caused by protease inhibitors from Avena sativa and Triticum durum to human macrophage-like cells. Macrophages were exposed (3 h) to extracts obtained from flours, and mitochondrial-associated oxygen consumption rates and inflammatory, metabolic, and proteome adaptations were quantified. Mass spectrometry ‘m/z’ signals of the extracts obtained from T. durum and A. sativa revealed molecular weights of 18–35 kDa and 16–22 kDa, respectively, for the compounds present at highest concentrations. Extracts from T. durum exhibited lower susceptibility to degradation by gastrointestinal enzymes than those from A. sativa: 9.5% vs 20.2%. Despite their different botanical origin, both extracts increased TLR4 expression. Metabolic protein levels were indicative of a decreased glycolytic to lactate flux in cell cultures upon stimulation with A. sativa extracts, which improved mitochondrial respiration in relation to those from T. durum. Principal components analysis confirmed relative similarities between immune–metabolic events triggered by immunonutritional ingredients in T. durum and A. sativa. Collectively, immunonutritional effects help to interpret the differences between both crops, worsening or improving, macrophage immune reactivity (tolerogenicity), and better control of inflammatory processes.
Collapse
|
11
|
Cronan MR, Hughes EJ, Brewer WJ, Viswanathan G, Hunt EG, Singh B, Mehra S, Oehlers SH, Gregory SG, Kaushal D, Tobin DM. A non-canonical type 2 immune response coordinates tuberculous granuloma formation and epithelialization. Cell 2021; 184:1757-1774.e14. [PMID: 33761328 PMCID: PMC8055144 DOI: 10.1016/j.cell.2021.02.046] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 11/03/2020] [Accepted: 02/22/2021] [Indexed: 12/19/2022]
Abstract
The central pathogen-immune interface in tuberculosis is the granuloma, a complex host immune structure that dictates infection trajectory and physiology. Granuloma macrophages undergo a dramatic transition in which entire epithelial modules are induced and define granuloma architecture. In tuberculosis, relatively little is known about the host signals that trigger this transition. Using the zebrafish-Mycobacterium marinum model, we identify the basis of granuloma macrophage transformation. Single-cell RNA-sequencing analysis of zebrafish granulomas and analysis of Mycobacterium tuberculosis-infected macaques reveal that, even in the presence of robust type 1 immune responses, countervailing type 2 signals associate with macrophage epithelialization. We find that type 2 immune signaling, mediated via stat6, is absolutely required for epithelialization and granuloma formation. In mixed chimeras, stat6 acts cell autonomously within macrophages, where it is required for epithelioid transformation and incorporation into necrotic granulomas. These findings establish the signaling pathway that produces the hallmark structure of mycobacterial infection.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified/genetics
- Animals, Genetically Modified/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Cell Differentiation
- Disease Models, Animal
- Epithelioid Cells/cytology
- Epithelioid Cells/immunology
- Epithelioid Cells/metabolism
- Granuloma/immunology
- Granuloma/metabolism
- Granuloma/pathology
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/metabolism
- Immunity/physiology
- Interferon-gamma/metabolism
- Interleukin-12/metabolism
- Macrophages/cytology
- Macrophages/immunology
- Macrophages/metabolism
- Mycobacterium Infections, Nontuberculous/immunology
- Mycobacterium Infections, Nontuberculous/pathology
- Mycobacterium marinum/isolation & purification
- Mycobacterium marinum/physiology
- Necrosis
- Receptors, Interleukin-4/antagonists & inhibitors
- Receptors, Interleukin-4/genetics
- Receptors, Interleukin-4/metabolism
- STAT6 Transcription Factor/antagonists & inhibitors
- STAT6 Transcription Factor/genetics
- STAT6 Transcription Factor/metabolism
- Signal Transduction
- Zebrafish/growth & development
- Zebrafish/metabolism
- RNA, Guide, CRISPR-Cas Systems
Collapse
Affiliation(s)
- Mark R Cronan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Erika J Hughes
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; University Program in Genetics and Genomics, Duke University School of Medicine, Durham, NC 27710, USA
| | - W Jared Brewer
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gopinath Viswanathan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Emily G Hunt
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Bindu Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia; The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW, Australia
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
12
|
Giorgio S, Gallo-Francisco PH, Roque GAS, Flóro E Silva M. Granulomas in parasitic diseases: the good and the bad. Parasitol Res 2020; 119:3165-3180. [PMID: 32789534 DOI: 10.1007/s00436-020-06841-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/02/2020] [Indexed: 12/12/2022]
Abstract
Parasitic diseases affect more than one billion people worldwide, and most of them are chronic conditions in which the treatment and prevention are difficult. The appearance of granulomas, defined as organized and compact structures of macrophages and other immune cells, during various parasitic diseases is frequent, since these structures will only form when individual immune cells do not control the invading agent. Th2-typering various parasitic diseases are frequent, since these structures will only form when individual immune cells do not control the invading agent. The characterization of granulomas in different parasitic diseases, as well as recent findings in this field, is discussed in this review, in order to understand the significance of the granuloma and its modulation in the host-parasite interaction and in the immune, pathological, and parasitological aspects of this interaction. The parasitic granulomatous diseases granulomatous amebic encephalitis, toxoplasmosis, leishmaniasis, neurocysticercosis, and schistosomiasis mansoni are discussed as well as the mechanistic and dynamical aspects of the infectious granulomas.
Collapse
Affiliation(s)
- Selma Giorgio
- Department of Animal Biology, Biology Institute, State University of Campinas, Campinas, 13083-865, Brazil.
| | | | | | - Marina Flóro E Silva
- Department of Animal Biology, Biology Institute, State University of Campinas, Campinas, 13083-865, Brazil
| |
Collapse
|
13
|
Sommariva M, Gagliano N. E-Cadherin in Pancreatic Ductal Adenocarcinoma: A Multifaceted Actor during EMT. Cells 2020; 9:E1040. [PMID: 32331358 PMCID: PMC7226001 DOI: 10.3390/cells9041040] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a step-wise process observed in normal and tumor cells leading to a switch from epithelial to mesenchymal phenotype. In tumors, EMT provides cancer cells with a metastatic phenotype characterized by E-cadherin down-regulation, cytoskeleton reorganization, motile and invasive potential. E-cadherin down-regulation is known as a key event during EMT. However, E-cadherin expression can be influenced by the different experimental settings and environmental stimuli so that the paradigm of EMT based on the loss of E-cadherin determining tumor cell behavior and fate often becomes an open question. In this review, we aimed at focusing on some critical points in order to improve the knowledge of the dynamic role of epithelial cells plasticity in EMT and, specifically, address the role of E-cadherin as a marker for the EMT axis.
Collapse
Affiliation(s)
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy;
| |
Collapse
|
14
|
Mukherjee S, Darzi S, Paul K, Cousins FL, Werkmeister JA, Gargett CE. Electrospun Nanofiber Meshes With Endometrial MSCs Modulate Foreign Body Response by Increased Angiogenesis, Matrix Synthesis, and Anti-Inflammatory Gene Expression in Mice: Implication in Pelvic Floor. Front Pharmacol 2020; 11:353. [PMID: 32265721 PMCID: PMC7107042 DOI: 10.3389/fphar.2020.00353] [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: 10/30/2019] [Accepted: 03/09/2020] [Indexed: 12/31/2022] Open
Abstract
Purpose Transvaginal meshes for the treatment of Pelvic Organ Prolapse (POP) have been associated with severe adverse events and have been banned for clinical use in many countries. We recently reported the design of degradable poly L-lactic acid-co-poly ε-caprolactone nanofibrous mesh (P nanomesh) bioengineered with endometrial mesenchymal stem/stromal cells (eMSC) for POP repair. We showed that such bioengineered meshes had high tissue integration as well as immunomodulatory effects in vivo. This study aimed to determine the key molecular players enabling eMSC-based foreign body response modulation. Methods SUSD2+ eMSC were purified from single cell suspensions obtained from endometrial biopsies from cycling women by magnetic bead sorting. Electrospun P nanomeshes with and without eMSC were implanted in a NSG mouse skin wound repair model for 1 and 6 weeks. Quantitative PCR was used to assess the expression of extracellular matrix (ECM), cell adhesion, angiogenesis and inflammation genes as log2 fold changes compared to sham controls. Histology and immunostaining were used to visualize the ECM, blood vessels, and multinucleated foreign body giant cells around implants. Results Bioengineered P nanomesh/eMSC constructs explanted after 6 weeks showed significant increase in 35 genes associated with ECM, ECM regulation, cell adhesion angiogenesis, and immune response in comparison to P nanomesh alone. In the absence of eMSC, acute inflammatory genes were significantly elevated at 1 week. However, in the presence of eMSC, there was an increased expression of anti-inflammatory genes including Mrc1 and Arg1 by 6 weeks. There was formation of multinucleated foreign body giant cells around both implants at 6 weeks that expressed CD206, a M2 macrophage marker. Conclusion This study reveals that eMSC modulate the foreign body response to degradable P nanomeshes in vivo by altering the expression profile of mouse genes. eMSC reduce acute inflammatory and increase ECM synthesis, angiogenesis and anti-inflammatory gene expression at 6 weeks while forming newly synthesized collagen within the nanomeshes and neo-vasculature in close proximity. From a tissue engineering perspective, this is a hallmark of a highly successful implant, suggesting significant potential as alternative surgical constructs for the treatment of POP.
Collapse
Affiliation(s)
- Shayanti Mukherjee
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Kallyanashis Paul
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Fiona L Cousins
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Jerome A Werkmeister
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
15
|
Yu W, Yang L, Li T, Zhang Y. Cadherin Signaling in Cancer: Its Functions and Role as a Therapeutic Target. Front Oncol 2019; 9:989. [PMID: 31637214 PMCID: PMC6788064 DOI: 10.3389/fonc.2019.00989] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022] Open
Abstract
Cadherin family includes lists of transmembrane glycoproteins which mediate calcium-dependent cell-cell adhesion. Cadherin-mediated adhesion regulates cell growth and differentiation throughout life. Through the establishment of the cadherin-catenin complex, cadherins provide normal cell-cell adhesion and maintain homeostatic tissue architecture. In the process of cell recognition and adhesion, cadherins act as vital participators. As results, the disruption of cadherin signaling has significant implications on tumor formation and progression. Altered cadherin expression plays a vital role in tumorigenesis, tumor progression, angiogenesis, and tumor immune response. Based on ongoing research into the role of cadherin signaling in malignant tumors, cadherins are now being considered as potential targets for cancer therapies. This review will demonstrate the mechanisms of cadherin involvement in tumor progression, and consider the clinical significance of cadherins as therapeutic targets.
Collapse
Affiliation(s)
- Weina Yu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Li Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Ting Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
Tavakolian S, Goudarzi H, Eslami G, Faghihloo E. Transcriptional Regulation of Epithelial to Mesenchymal Transition Related Genes by Lipopolysaccharide in Human Cervical Cancer Cell Line HeLa. Asian Pac J Cancer Prev 2019; 20:2455-2461. [PMID: 31450920 PMCID: PMC6852821 DOI: 10.31557/apjcp.2019.20.8.2455] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Indexed: 12/29/2022] Open
Abstract
Objective: Cancer is one of the common diseases in the world, and cervical cancer is the fourth one. In this type of cancer, many risk factors, especially infectious diseases, such as human papilloma virus (HPV) and gram-negative bacteria can have important effects on the expression of epithelial to mesenchymal transition related genes like Snail,E-cadherin, and ZEB-1, responsible for connecting cell tissues. In this study, we have investigated the effect of Escherichia coli O111:B4 Lipopolysaccharide (LPS) on HPV positive cell line (HeLa), the expression level of the (Snail, E-cadherin, and ZEB-1), HPV oncogenes (E6, E7) and also microRNA-9, 192. Materials and Methods: HeLa cell line was treated with LPS to analyze Snail, E-cadherin, ZEB-1, E6, E7 and also microRNA-9, 192 expression by quantitative real-time PCR in 24, 48 and 72 hours. Results: Quantitative real-time PCR revealed a significant reduction in E-cadherin mRNA level at 10ug/L of LPS in three time-points and after 24 hours at 5ug/L of LPS; however, ZEB-1 at 10ug/L of LPS and Snail at 5, 10ug/L of LPS are up-regulated. E7 also illustrated a slight increase, but we did not find any relationship between E7 and LPS treatment. Additionally, there are upward trends in microRNA-9, 192 levels. Conclusion: The result of this study, LPS is able to reduce E-cadherin expression, caused by increase in repressor E-cadherin protein expression and some microRNAs, probably. Since bacterial infection can be in cervical site, it is likely to be effective in reducing the E-cadherin expression in the EMT and enhance cancer process, therefore; removing these infections by using the appropriate antibiotics may result in slowing down this process, which requires more research.
Collapse
Affiliation(s)
- Shaian Tavakolian
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hossein Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Gita Eslami
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ebrahim Faghihloo
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Tu MG, Sun KT, Wang TH, He YZ, Hsia SM, Tsai BH, Shih YH, Shieh TM. Effects of mineral trioxide aggregate and bioceramics on macrophage differentiation and polarization in vitro. J Formos Med Assoc 2019; 118:1458-1465. [PMID: 31358435 DOI: 10.1016/j.jfma.2019.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/04/2019] [Accepted: 07/11/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND/PURPOSE Mineral trioxide aggregate (Pro-Root MTA, PR-MTA) and bioceramics (iRoot® SP Injectable Root Canal Sealer, iR-BC) are used for making apical plugs used in apexification, repairing root perforations during root canal therapy, and treating internal root resorption. The purpose of the present in vitro study was to compare the biological effects of PR-MTA- and iR-BC-based dental sealers in the mouse macrophage cell line RAW 264.7. METHODS Cytotoxicity and cell proliferation were analyzed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and cell hemocytometer, respectively. Protein expression of biomarkers of cell proliferation, autophagy, and osteoclast differentiation was determined by western blotting. Pro-inflammatory gene expression was examined using quantitative reverse transcription-PCR. RESULTS PR-MTA induced cytotoxicity in RAW 264.7 cells in a dose-dependent manner, and iR-BC was more cytotoxic than PR-MTA. Low-dose and short-term treatments of both PR-MTA and iR-BC induced RAW 264.7 cell proliferation. PR-MTA induced autophagy, whereas iR-BC did not. Neither PR-MTA nor iR-BC induced osteoclastogenesis. Pro-inflammatory genes were activated by both materials. However, the expression of inducible nitric oxide synthase (iNOS) mRNA was upregulated by iR-BC treatment, but not by PR-MTA treatment. CONCLUSION Overall, dental PR-MTA and iR-BC induced pro-inflammatory genes but did not induce osteoclastogenesis in macrophages. PR-MTA and iR-BC induced M2 and M1 polarization, respectively, of RAW 264.7 cells.
Collapse
Affiliation(s)
- Ming-Gene Tu
- School of Dentistry, China Medical University, Taichung, Taiwan
| | - Kuo-Ting Sun
- School of Dentistry, China Medical University, Taichung, Taiwan; Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan; Department of Pediatric Dentistry, China Medical University Hospital, Taichung, Taiwan
| | - Tong-Hong Wang
- Tissue Bank, Chang Gung Memorial Hospital, Linko, Taiwan
| | - Yun-Zhen He
- School of Dentistry, China Medical University, Taichung, Taiwan
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Bi-He Tsai
- Department of Dental Hygiene, China Medical University, Taichung, Taiwan
| | - Yin-Hwa Shih
- Department of Healthcare Administration, Asia University, Taichung, Taiwan.
| | - Tzong-Ming Shieh
- Department of Dental Hygiene, China Medical University, Taichung, Taiwan.
| |
Collapse
|
18
|
Brooks PJ, Glogauer M, McCulloch CA. An Overview of the Derivation and Function of Multinucleated Giant Cells and Their Role in Pathologic Processes. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1145-1158. [PMID: 30926333 DOI: 10.1016/j.ajpath.2019.02.006] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/04/2019] [Accepted: 02/13/2019] [Indexed: 12/20/2022]
Abstract
Monocyte lineage cells play important roles in health and disease. Their differentiation into macrophages is crucial for a broad array of immunologic processes that regulate inflammation, neoplasia, and infection. In certain pathologic conditions, such as foreign body reactions and peripheral inflammatory lesions, monocytes fuse to form large, multinucleated giant cells (MGCs). Currently, our knowledge of the fusion mechanisms of monocytes and the regulation of MGC formation and function in discrete pathologies is limited. Herein, we consider the types and function of MGCs in disease and assess the mechanisms by which monocyte fusion contributes to the formation of MGCs. An improved understanding of the cellular origins and metabolic functions of MGCs will facilitate their identification and ultimately the treatment of diseases and disorders that involve MGCs.
Collapse
Affiliation(s)
- Patricia J Brooks
- Matrix Dynamics Group, University of Toronto, Toronto, Ontario, Canada; Department of Dental Oncology and Maxillofacial Prosthetics, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.
| | - Michael Glogauer
- Matrix Dynamics Group, University of Toronto, Toronto, Ontario, Canada; Department of Dental Oncology and Maxillofacial Prosthetics, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | |
Collapse
|
19
|
Longo V, Longo A, Di Sano C, Cigna D, Cibella F, Di Felice G, Colombo P. In vitro exposure to 2,2',4,4'-tetrabromodiphenyl ether (PBDE-47) impairs innate inflammatory response. CHEMOSPHERE 2019; 219:845-854. [PMID: 30562690 DOI: 10.1016/j.chemosphere.2018.12.082] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/03/2018] [Accepted: 12/10/2018] [Indexed: 06/09/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) are persistent organic pollutants that are added to numerous products to prevent accidental fires. PBDEs are present in the environment and they bio-accumulate in human and animal tissues. Recently, their presence has been correlated to several pathologies but little is known about their effect on the human innate immune system activity. In this study we investigated the effect of the congener 2,2',4,4'-Tetrabromodiphenyl ether (PBDE-47) on the functional activity of the THP-1 human macrophages cell line and on ex vivo freshly isolated human basophils. Cytotoxicity and genotoxicity studies showed that PBDE-47 was able to induce toxic effects on the THP-1 cell line viability at concentrations ≥25 μM. Immune function of THP-1 was studied after stimulation with bacterial lipopolysaccharide (LPS) and PBDE-47 exposure at concentrations granting macrophage viability. Two dimensional electrophoresis showed modification of the proteome in the 3 μM PBDE-47 treated sample and Real Time PCR and ELISA demonstrated a statistically significant reduction in the expression of IL-1β, IL-6 and TNF-α cytokines. Furthermore, PBDE-47 was able to perturbate genes involved in cell motility upregulating CDH-1 and downregulating MMP-12 expressions. Finally, basophil activation assay showed reduced CD63 activation in PBDE-47 treated samples. In conclusion, our study demonstrated that PBDE-47 may perturb the activities of cells involved in innate immunity dampening the expression of macrophage pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α) and genes involved in cell motility (MMP-12 and E-cadherin) and interfering with basophil activation suggesting that this compound can impair innate immune response.
Collapse
Affiliation(s)
- Valeria Longo
- Istituto di Biomedicina e di Immunologia Molecolare del Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Alessandra Longo
- Istituto di Biomedicina e di Immunologia Molecolare del Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Caterina Di Sano
- Istituto di Biomedicina e di Immunologia Molecolare del Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Diego Cigna
- Istituto di Biomedicina e di Immunologia Molecolare del Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Fabio Cibella
- Istituto di Biomedicina e di Immunologia Molecolare del Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Gabriella Di Felice
- Istituto Superiore di Sanità, National Center for Drug Research and Evaluation, Rome, Italy
| | - Paolo Colombo
- Istituto di Biomedicina e di Immunologia Molecolare del Consiglio Nazionale delle Ricerche, Palermo, Italy.
| |
Collapse
|
20
|
Jiang M, Zhang J, Qian L, Miao Y, Song W, Liu H, Li R. MOZ Forms an Autoregulatory Feedback Loop with miR-223 in AML and Monocyte/Macrophage Development. iScience 2019; 11:189-204. [PMID: 30616103 PMCID: PMC6321978 DOI: 10.1016/j.isci.2018.12.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 09/11/2018] [Accepted: 12/18/2018] [Indexed: 11/22/2022] Open
Abstract
Monocytic leukemia zinc-finger protein (MOZ) has been found to form fusion proteins with many regulators in acute myeloid leukemia (AML). However, the molecular functions and underlying mechanism of MOZ in AML is not well understood. Here, clinical MOZ expression analysis combined with data integration from the TCGA and GEO databases indicated that a low level of MOZ was associated with poor prognosis. MOZ knockdown inhibited monocyte differentiation and increased resistance to chemotherapeutic drug-induced apoptosis in THP-1 or U937 cells. In addition, we found that genetic silencing of MOZ suppressed AP-1 and AKT activity in the context of lipopolysaccharide stimulation, resulting in diminished M1 activation of macrophages. We further showed that MOZ was a validated target of miR-223 and functioned as a repressor of miR-223 expression. Our study indicates that a molecular network involving MOZ and miR-223 contributes to the monocyte differentiation and polarization program, which is deregulated in AML.
Collapse
Affiliation(s)
- Ming Jiang
- School of Life Science, University of Science and Technology of China, Hefei 230027, Anhui, China.
| | - Ju Zhang
- Department of Laboratory Medicine, Anhui Provincial Hospital, Hefei 230001, Anhui, China; Department of Laboratory Medicine, First Affiliated Hospital of University of Science and Technology of China, Hefei 230001, Anhui, China
| | - Lili Qian
- Department of Laboratory Medicine, Anhui Provincial Hospital, Hefei 230001, Anhui, China
| | - Yuhui Miao
- School of Life Science, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Weiguo Song
- Department of Laboratory Medicine, Anhui Provincial Hospital, Hefei 230001, Anhui, China
| | - Hanyuan Liu
- Department of Laboratory Medicine, Anhui Provincial Hospital, Hefei 230001, Anhui, China
| | - Rui Li
- School of Life Science, University of Science and Technology of China, Hefei 230027, Anhui, China
| |
Collapse
|
21
|
Gao L, Liu MM, Zang HM, Ma QY, Yang Q, Jiang L, Ren GL, Li HD, Wu WF, Wang JN, Wei B, Liu XQ, Jiang C, Huang C, Li J, Meng XM. Restoration of E-cadherin by PPBICA protects against cisplatin-induced acute kidney injury by attenuating inflammation and programmed cell death. J Transl Med 2018; 98:911-923. [PMID: 29581579 DOI: 10.1038/s41374-018-0052-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 02/04/2018] [Accepted: 02/07/2018] [Indexed: 12/22/2022] Open
Abstract
E-cadherin is a major component of tubular adherent proteins that maintain intercellular contacts and cell polarity in epithelial tissue. It is involved in pathological processes of renal cell carcinoma and fibrotic diseases via epithelial-mesenchymal transition. Although studies have shown E-cadherin is significantly downregulated in acute kidney injury (AKI), its function in AKI is unknown. Here, we evaluated cell damage and inflammation in cisplatin-stimulated tubular epithelial cell lines after disrupting E-cadherin and restoring it with PPBICA, a small molecule identified by high-throughput screening. We also determined the therapeutic potential of restoring E-cadherin in vivo. Results show cisplatin reduced E-cadherin expression both in mouse kidney and proximal tubular epithelial cell lines (mTECs). PPBICA restored E-cadherin levels, which increased cell viability while attenuating programmed cell death. This may be mediated via deactivation of the RIPK1/RIPK3 axis and decreased caspase3 cleavage. In addition, PPBICA suppressed inflammatory response in cisplatin-treated mTECs, which correlated with suppressed NF-κB phosphorylation and promoter activity. In contrast, disruption of E-cadherin promoted cell damage and inflammation. PPBICA failed to further attenuate kidney damage in E-cadherin knockdown cells, indicating that PPBICA protects against mTECs through E-cadherin restoration. We also found that peritoneal injection of PPBICA in mice prevented loss of renal function and tubular damage by suppressing NF-κB-driven renal inflammation and RIPK-regulated programmed cell death. This was driven by restoration of E-cadherin in cisplatin nephropathy. Additionally, PPBICA attenuated cisplatin-induced kidney damage in an established AKI model, indicating its therapeutic potential in the treatment of AKI. In conclusion, E-cadherin plays functional roles in tubule integrity, programmed cell death, and renal inflammation. Our results underscore the potential of E-cadherin restoration as a novel therapeutic strategy for AKI.
Collapse
Affiliation(s)
- Li Gao
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Ming-Ming Liu
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Hong-Mei Zang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China.,Anhui Institute of Innovative Drugs, Hefei, Anhui, China.,Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui, 230032, China
| | - Qiu-Ying Ma
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Qin Yang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Ling Jiang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Gui-Ling Ren
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Hai-Di Li
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Wei-Feng Wu
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Jia-Nan Wang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Biao Wei
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Xue-Qi Liu
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Cheng Jiang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Cheng Huang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China.,Anhui Institute of Innovative Drugs, Hefei, Anhui, China.,Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui, 230032, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China.,Anhui Institute of Innovative Drugs, Hefei, Anhui, China.,Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui, 230032, China
| | - Xiao-Ming Meng
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, China. .,Anhui Institute of Innovative Drugs, Hefei, Anhui, China. .,Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Hefei, Anhui, 230032, China.
| |
Collapse
|
22
|
Chiarella SE, Rabin EE, Ostilla LA, Flozak AS, Gottardi CJ. αT-catenin: A developmentally dispensable, disease-linked member of the α-catenin family. Tissue Barriers 2018; 6:e1463896. [PMID: 29746206 PMCID: PMC6179130 DOI: 10.1080/21688370.2018.1463896] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/19/2018] [Accepted: 02/23/2018] [Indexed: 02/07/2023] Open
Abstract
α-Catenins are actin-filament binding proteins and critical subunits of the cadherin-catenin cell-cell adhesive complex. They are found in nominally-defined epithelial (E), neural (N), and testis (T) forms transcribed from three distinct genes. While most of α-catenin research has focused on the developmentally essential founding member, αE-catenin, this review discusses recent studies on αT-catenin (CTNNA3), a developmentally dispensable isoform that is emerging as relevant to cardiac, allergic and neurological diseases.
Collapse
Affiliation(s)
- Sergio E. Chiarella
- Department of Medicine
- Cellular and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Erik E. Rabin
- Department of Medicine
- Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL
| | - Lorena A. Ostilla
- Department of Medicine
- Cellular and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Annette S. Flozak
- Department of Medicine
- Cellular and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL
| | - Cara J. Gottardi
- Department of Medicine
- Cellular and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
23
|
Díaz Á, Sagasti C, Casaravilla C. Granulomatous responses in larval taeniid infections. Parasite Immunol 2018. [DOI: 10.1111/pim.12523] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Á. Díaz
- Área/Cátedra de Inmunología; Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias); Universidad de la República; Montevideo Uruguay
| | - C. Sagasti
- Área/Cátedra de Inmunología; Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias); Universidad de la República; Montevideo Uruguay
| | - C. Casaravilla
- Área/Cátedra de Inmunología; Departamento de Biociencias (Facultad de Química) e Instituto de Química Biológica (Facultad de Ciencias); Universidad de la República; Montevideo Uruguay
| |
Collapse
|
24
|
Fontenete S, Peña-Jimenez D, Perez-Moreno M. Heterocellular cadherin connections: coordinating adhesive cues in homeostasis and cancer. F1000Res 2017; 6:1010. [PMID: 28721207 PMCID: PMC5497824 DOI: 10.12688/f1000research.11357.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/26/2017] [Indexed: 01/06/2023] Open
Abstract
This short insight covers some of the recent topics relevant to the field of cadherin-catenin adhesion in mediating connections between different cell types, so-called heterotypic or heterocellular connections, in both homeostasis and cancer. These scientific discoveries are increasing our understanding of how multiple cells residing in complex tissues can be instructed by cadherin adhesion receptors to regulate tissue architecture and function and how these cadherin-mediated heterocellular connections spur tumor growth and the acquisition of malignant characteristics in tumor cells. Overall, the findings that have emerged over the past few years are elucidating the complexity of the functional roles of the cadherin-catenin complexes. Future exciting research lies ahead in order to understand the physical basis of these heterotypic interactions and their influence on the behavior of heterogeneous cellular populations as well as their roles in mediating phenotypic and genetic changes as cells evolve through complex environments during morphogenesis and cancer.
Collapse
Affiliation(s)
- Silvia Fontenete
- Epithelial Cell Biology Group, Cancer Cell Biology Programme, Spanish Cancer Research Centre (CNIO), Madrid, Spain
| | - Daniel Peña-Jimenez
- Epithelial Cell Biology Group, Cancer Cell Biology Programme, Spanish Cancer Research Centre (CNIO), Madrid, Spain
| | - Mirna Perez-Moreno
- Epithelial Cell Biology Group, Cancer Cell Biology Programme, Spanish Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
25
|
Albright JM, Dunn RC, Shults JA, Boe DM, Afshar M, Kovacs EJ. Advanced Age Alters Monocyte and Macrophage Responses. Antioxid Redox Signal 2016; 25:805-815. [PMID: 27357201 PMCID: PMC5107740 DOI: 10.1089/ars.2016.6691] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
SIGNIFICANCE With the growing population of baby boomers, there is a great need to determine the effects of advanced age on the function of the immune system. Recent Advances: It is universally accepted that advanced age is associated with a chronic low-grade inflammatory state that is referred to as inflamm-aging, which alters the function of both immune and nonimmune cells. Mononuclear phagocytes play a central role in both the initiation and resolution of inflammation in multiple organ systems and exhibit marked changes in phenotype and function in response to environmental cues, including the low levels of pro-inflammatory mediators seen in the aged. CRITICAL ISSUES Although we know a great deal about the function of immune cells in young adults and there is a growing body of literature focusing on aging of the adaptive immune system, much less is known about the impact of age on innate immunity and the critical role of the mononuclear phagocytes in this process. FUTURE DIRECTIONS In this article, there is a focus on the tissue-specific monocyte and macrophage subsets and how they are altered in the aged milieu, with the hope that this compilation of observations will spark an expansion of research in the field. Antioxid. Redox Signal. 25, 805-815.
Collapse
Affiliation(s)
- Joslyn M Albright
- 1 Department of Surgery, Loyola University Chicago Health Sciences Campus , Maywood, Illinois.,2 Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Campus , Maywood, Illinois
| | - Robert C Dunn
- 2 Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Campus , Maywood, Illinois.,3 Stritch School of Medicine, Loyola University Chicago Health Sciences Campus , Maywood, Illinois
| | - Jill A Shults
- 1 Department of Surgery, Loyola University Chicago Health Sciences Campus , Maywood, Illinois.,2 Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Campus , Maywood, Illinois
| | - Devin M Boe
- 4 Department of Surgery, University of Colorado Denver Anschutz Medical Campus , Aurora, Colorado
| | - Majid Afshar
- 2 Burn and Shock Trauma Research Institute, Loyola University Chicago Health Sciences Campus , Maywood, Illinois.,3 Stritch School of Medicine, Loyola University Chicago Health Sciences Campus , Maywood, Illinois.,5 Department of Medicine, Loyola University Chicago Health Sciences Campus , Maywood, Illinois.,6 Department of Public Health Sciences, Loyola University Chicago Health Sciences Campus , Maywood, Illinois
| | - Elizabeth J Kovacs
- 4 Department of Surgery, University of Colorado Denver Anschutz Medical Campus , Aurora, Colorado
| |
Collapse
|
26
|
Cronan MR, Beerman RW, Rosenberg AF, Saelens JW, Johnson MG, Oehlers SH, Sisk DM, Jurcic Smith KL, Medvitz NA, Miller SE, Trinh LA, Fraser SE, Madden JF, Turner J, Stout JE, Lee S, Tobin DM. Macrophage Epithelial Reprogramming Underlies Mycobacterial Granuloma Formation and Promotes Infection. Immunity 2016; 45:861-876. [PMID: 27760340 PMCID: PMC5268069 DOI: 10.1016/j.immuni.2016.09.014] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 07/21/2016] [Accepted: 07/29/2016] [Indexed: 01/23/2023]
Abstract
Mycobacterium tuberculosis infection in humans triggers formation of granulomas, which are tightly organized immune cell aggregates that are the central structure of tuberculosis. Infected and uninfected macrophages interdigitate, assuming an altered, flattened appearance. Although pathologists have described these changes for over a century, the molecular and cellular programs underlying this transition are unclear. Here, using the zebrafish-Mycobacterium marinum model, we found that mycobacterial granuloma formation is accompanied by macrophage induction of canonical epithelial molecules and structures. We identified fundamental macrophage reprogramming events that parallel E-cadherin-dependent mesenchymal-epithelial transitions. Macrophage-specific disruption of E-cadherin function resulted in disordered granuloma formation, enhanced immune cell access, decreased bacterial burden, and increased host survival, suggesting that the granuloma can also serve a bacteria-protective role. Granuloma macrophages in humans with tuberculosis were similarly transformed. Thus, during mycobacterial infection, granuloma macrophages are broadly reprogrammed by epithelial modules, and this reprogramming alters the trajectory of infection and the associated immune response.
Collapse
Affiliation(s)
- Mark R Cronan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Rebecca W Beerman
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Allison F Rosenberg
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Joseph W Saelens
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Matthew G Johnson
- Division of Infectious Diseases, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Stefan H Oehlers
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Dana M Sisk
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kristen L Jurcic Smith
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Neil A Medvitz
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sara E Miller
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Le A Trinh
- Molecular and Computational Biology and Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA
| | - Scott E Fraser
- Molecular and Computational Biology and Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA
| | - John F Madden
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Joanne Turner
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Center for Microbial Interface Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Jason E Stout
- Division of Infectious Diseases, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Sunhee Lee
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
27
|
Fiorino C, Harrison RE. E-cadherin is important for cell differentiation during osteoclastogenesis. Bone 2016; 86:106-18. [PMID: 26959175 DOI: 10.1016/j.bone.2016.03.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/29/2016] [Accepted: 03/04/2016] [Indexed: 01/05/2023]
Abstract
E-cadherin, a protein responsible for intercellular adhesion between epithelial cells, is also expressed in the monocyte/macrophage lineage. In this study we have explored the involvement of E-cadherin during receptor activator of nuclear factor-κB ligand (RANKL)-stimulated osteoclast differentiation. Osteoclastogenesis involves a period of precursor expansion followed by multiple fusion events to generate a multinuclear osteoclast that is capable of bone resorption. We asked whether E-cadherin participated in early precursor interactions and recognition or was a component of the osteoclast fusion machinery. Here, we show that endogenous E-cadherin expression is the highest during early stages of osteoclast differentiation, with surface expression visible on small precursor cells (fewer than four nuclei per cell) in both RAW 264.7 cells and primary macrophages. Blocking E-cadherin function with neutralizing antibodies prior to the onset of fusion delayed the expression of TRAP, Cathepsin K, DC-STAMP and NFATc1 and significantly diminished multinucleated osteoclast formation. Conversely, E-cadherin-GFP overexpressing macrophages displayed earlier NFATc1 nuclear translocation along with faster formation of multinucleated osteoclasts compared to control macrophages. Through live imaging we identified that disrupting E-cadherin function prolonged the proliferative phase of the precursor population while concomitantly decreasing the proportion of migrating precursors. The lamellipodium and polarized membrane extensions appeared to be the principal sites of fusion, indicating precursor migration was a critical factor contributing to osteoclast fusion. These findings demonstrate that E-cadherin-mediated cell-cell contacts can modulate osteoclast-specific gene expression and prompt differentiating osteoclast precursors toward migratory and fusion activities.
Collapse
Affiliation(s)
- Cara Fiorino
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario M1C 1A4, Canada; Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada
| | - Rene E Harrison
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario M1C 1A4, Canada; Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario M1C 1A4, Canada.
| |
Collapse
|
28
|
e-Cadherin in 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-Induced Parkinson Disease. Mediators Inflamm 2016; 2016:3937057. [PMID: 27194825 PMCID: PMC4852763 DOI: 10.1155/2016/3937057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/29/2016] [Indexed: 12/11/2022] Open
Abstract
Today a large number of studies are focused on clarifying the complexity and diversity of the pathogenetic mechanisms inducing Parkinson disease. We used 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), a neurotoxin that induces Parkinson disease, to evaluate the change of midbrain structure and the behavior of the anti-inflammatory factor e-cadherin, interleukin-6, tyrosine hydroxylase, phosphatase and tensin homolog, and caveolin-1. The results showed a strong expression of e-cadherin, variation of length and thickness of the heavy neurofilaments, increase of interleukin-6, and reduction of tyrosine hydroxylase known to be expression of dopamine cell loss, reduction of phosphatase and tensin homolog described to impair responses to dopamine, and reduction of caveolin-1 known to be expression of epithelial-mesenchymal transition and fibrosis. The possibility that the overexpression of the e-cadherin might be implicated in the anti-inflammatory reaction to MPTP treatment by influencing the behavior of the other analyzed molecules is discussed.
Collapse
|