201
|
Toll-Like Receptor 2 Expression as a New Hallmark of Advanced Endometriosis. Cells 2020; 9:cells9081813. [PMID: 32751735 PMCID: PMC7464841 DOI: 10.3390/cells9081813] [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: 06/22/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/27/2022] Open
Abstract
Recent evidence suggests that immunological aspects play a pivotal role in this disorder. Toll-like receptor 2 (TLR2) is crucial in recognizing microbial infections and mediating innate immune response. The objective of our study was to rate with flow cytometry the levels of several subsets of dendritic cells, monocytes, and basic peripheral blood lymphocytes expressing TLR2, aiming at the determination of a possible correlation between the expression of TLR2 and the clinical outcomes of endometriosis in 40 patients and 40 age-matched healthy women. Our study showed the importance of TLR2 expression, mainly on myeloid dendritic cells (mDCs) and B cells in patients with endometriosis. Both mDCs BDCA1+CD19-TLR2+ and B lymphocytes CD19+TLR-2+ proved useful in the differentiation of affected individuals with stages 3–4 of the disease (area under the receiver operating characteristic curve /AUC/ = 0.96, p < 0.0001 for mDCs; AUC = 0.78, p = 0.0001 for B lymphocytes), and those presenting adhesion (AUC = 0.92, p < 0.0001 for mDCs; AUC = 0.82, p < 0.0001 for B lymphocytes) or infertility (AUC = 0.83, p < 0.0001 for mDCs; AUC = 0.73, p = 0.006 for B lymphocytes). Our findings suggest that the levels of TLR2-expressing cells, particularly mDCs and B lymphocytes, may be an effective biomarker of endometriosis, because the disease currently lacks clinically useful noninvasive biomarkers enabling early and cost-effective diagnosis.
Collapse
|
202
|
Ulbrich P, Khoshneviszadeh M, Jandke S, Schreiber S, Dityatev A. Interplay between perivascular and perineuronal extracellular matrix remodelling in neurological and psychiatric diseases. Eur J Neurosci 2020; 53:3811-3830. [DOI: 10.1111/ejn.14887] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/29/2020] [Accepted: 06/18/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Philipp Ulbrich
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
- Department of Neurology Otto‐von‐Guericke University Magdeburg Germany
| | - Mahsima Khoshneviszadeh
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
- Department of Neurology Otto‐von‐Guericke University Magdeburg Germany
| | - Solveig Jandke
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
- Department of Neurology Otto‐von‐Guericke University Magdeburg Germany
| | - Stefanie Schreiber
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
- Department of Neurology Otto‐von‐Guericke University Magdeburg Germany
- Center for Behavioral Brain Sciences (CBBS) Magdeburg Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE) Magdeburg Germany
- Center for Behavioral Brain Sciences (CBBS) Magdeburg Germany
- Medical Faculty Otto‐von‐Guericke University Magdeburg Germany
| |
Collapse
|
203
|
Strapcova S, Takacova M, Csaderova L, Martinelli P, Lukacikova L, Gal V, Kopacek J, Svastova E. Clinical and Pre-Clinical Evidence of Carbonic Anhydrase IX in Pancreatic Cancer and Its High Expression in Pre-Cancerous Lesions. Cancers (Basel) 2020; 12:E2005. [PMID: 32707920 PMCID: PMC7464147 DOI: 10.3390/cancers12082005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
Hypoxia is a common phenomenon that occurs in most solid tumors. Regardless of tumor origin, the evolution of a hypoxia-adapted phenotype is critical for invasive cancer development. Pancreatic ductal adenocarcinoma is also characterized by hypoxia, desmoplasia, and the presence of necrosis, predicting poor outcome. Carbonic anhydrase IX (CAIX) is one of the most strict hypoxia regulated genes which plays a key role in the adaptation of cancer cells to hypoxia and acidosis. Here, we summarize clinical data showing that CAIX expression is associated with tumor necrosis, vascularization, expression of Frizzled-1, mucins, or proteins involved in glycolysis, and inevitably, poor prognosis of pancreatic cancer patients. We also describe the transcriptional regulation of CAIX in relation to signaling pathways activated in pancreatic cancers. A large part deals with the preclinical evidence supporting the relevance of CAIX in processes leading to the aggressive behavior of pancreatic tumors. Furthermore, we focus on CAIX occurrence in pre-cancerous lesions, and for the first time, we describe CAIX expression within intraductal papillary mucinous neoplasia. Our review concludes with a detailed account of clinical trials implicating that treatment consisting of conventionally used therapies combined with CAIX targeting could result in an improved anti-cancer response in pancreatic cancer patients.
Collapse
Affiliation(s)
- Sabina Strapcova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| | - Martina Takacova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| | - Lucia Csaderova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| | - Paola Martinelli
- Institute of Cancer Research, Clinic of Internal Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
- Cancer Cell Signaling, Boehringer-Ingelheim RCV Vienna, A-1121 Vienna, Austria
| | - Lubomira Lukacikova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| | - Viliam Gal
- Alpha Medical Pathology, Ruzinovska 6, 82606 Bratislava, Slovakia;
| | - Juraj Kopacek
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| | - Eliska Svastova
- Department of Tumor Biology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia; (S.S.); (M.T.); (L.C.); (L.L.); (J.K.)
| |
Collapse
|
204
|
Giovannetti A, Straface E, Rosato E, Casciaro M, Pioggia G, Gangemi S. Role of Alarmins in the Pathogenesis of Systemic Sclerosis. Int J Mol Sci 2020; 21:ijms21144985. [PMID: 32679721 PMCID: PMC7404317 DOI: 10.3390/ijms21144985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 12/22/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare chronic autoimmune disease associated with significant morbidity and mortality. Two main subsets of SSc are recognized: (i) diffuse cutaneous SSc with rapidly progressive fibrosis of the skin, lungs, and other internal organs; and (ii) limited cutaneous SSc, which is dominated by vascular manifestations, with skin and organ fibrosis generally limited and slowly progressing. In spite of intense investigation, both etiology and pathogenesis of SSc are still unknown. Genetic and environmental factors, as well as abnormalities of immune functions, are strongly suggested for etiology, while microvascular abnormalities, immune system activation, and oxidative stress are suggested for the pathogenesis. Recently, it has been found that a multitude of mediators and cytokines are implicated in the fibrotic processes observed in SSc. Among these, a central role could be exerted by “alarmins”, endogenous and constitutively expressed proteins/peptides that function as an intercellular signal defense. This review describes, in a detailed manner, the role of alarmins in the pathogenesis of scleroderma.
Collapse
Affiliation(s)
- Antonello Giovannetti
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy;
- Correspondence: ; Tel.: +39-3476138512
| | - Elisabetta Straface
- Center for Gender-Specific Medicine, Biomarkers Unit, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Edoardo Rosato
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Marco Casciaro
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.C.); (S.G.)
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy; (M.C.); (S.G.)
| |
Collapse
|
205
|
Liao Y, Liu S, Fu S, Wu J. HMGB1 in Radiotherapy: A Two Headed Signal Regulating Tumor Radiosensitivity and Immunity. Onco Targets Ther 2020; 13:6859-6871. [PMID: 32764978 PMCID: PMC7369309 DOI: 10.2147/ott.s253772] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy (RT) is a mainstay of cancer treatment. Recent studies have shown that RT not only directly induces cell death but also has late and sustained immune effects. High mobility group box 1 (HMGB1) is a nuclear protein released during RT, with location-dependent functions. It is essential for normal cellular function but also regulates the proliferation and migration of tumor cells by binding to high-affinity receptors. In this review, we summarize recent evidence on the functions of HMGB1 in RT according to the position, intracellular HMGB1 and extracellular HMGB1. Intracellular HMGB1 induces radiation tolerance in tumor cells by promoting DNA damage repair and autophagy. Extracellular HMGB1 plays a more intricate role in radiation-related immune responses, wherein it not only stimulates the anti-tumor immune response by facilitating the recognition of dying tumor cells but is also involved in maintaining immunosuppression. Factors that potentially affect the role of HMGB1 in RT-induced cytotoxicity have also been discussed in the context of possible therapeutic applications, which helps to develop effective and targeted radio-sensitization therapies.
Collapse
Affiliation(s)
- Yin Liao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Shuya Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| |
Collapse
|
206
|
Abstract
PURPOSE OF REVIEW The innate immune system is essential in the protection against microbial infection and facilitating tissue repair mechanisms. During these stresses, the maintenance of innate immune cell numbers through stress-induced or emergency hematopoiesis is key for our survival. One major mechanism to recognize danger signals is through the activation of Toll-like receptors (TLRs) on the surface of hematopoietic cells, including hematopoietic stem cell (HSC) and hematopoietic progenitor cell (HPC), and nonhematopoietic cells, which recognize pathogen-derived or damaged-induced compounds and can influence the emergency hematopoietic response. This review explores how direct pathogen-sensing by HSC/HPC regulates hematopoiesis, and the positive and negative consequences of these signals. RECENT FINDINGS Recent studies have highlighted new roles for TLRs in regulating HSC and HPC differentiation to innate immune cells of both myeloid and lymphoid origin and augmenting HSC and HPC migration capabilities. Most interestingly, new insights as to how acute versus chronic stimulation of TLR signaling regulates HSC and HPC function has been explored. SUMMARY Recent evidence suggests that TLRs may play an important role in many inflammation-associated diseases. This suggests a possible use for TLR agonists or antagonists as potential therapeutics. Understanding the direct effects of TLR signaling by HSC and HPC may help regulate inflammatory/danger signal-driven emergency hematopoiesis.
Collapse
|
207
|
Requirements for the differentiation of innate T-bet high memory-phenotype CD4 + T lymphocytes under steady state. Nat Commun 2020; 11:3366. [PMID: 32632165 PMCID: PMC7338451 DOI: 10.1038/s41467-020-17136-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 06/11/2020] [Indexed: 12/13/2022] Open
Abstract
CD4+ T lymphocytes consist of naïve, antigen-specific memory, and memory-phenotype (MP) cell compartments at homeostasis. We recently showed that MP cells exert innate-like effector function during host defense, but whether MP CD4+ T cells are functionally heterogeneous and, if so, what signals specify the differentiation of MP cell subpopulations under homeostatic conditions is still unclear. Here we characterize MP lymphocytes as consisting of T-bethigh, T-betlow, and T-bet− subsets, with innate, Th1-like effector activity exclusively associated with T-bethigh cells. We further show that the latter population depends on IL-12 produced by CD8α+ type 1 dendritic cells (DC1) for its differentiation. Finally, our data demonstrate that this tonic IL-12 production requires TLR-MyD88 signaling independent of foreign agonists, and is further enhanced by CD40-CD40L interactions between DC1 and CD4+ T lymphocytes. We propose that optimal differentiation of T-bethigh MP lymphocytes at homeostasis is driven by self-recognition signals at both the DC and Tcell levels. CD4+ T cells contain a T-bethigh memory-phenotype (MP) population with innate-like functions. Here the authors characterize the requirements for their differentiation at homeostasis and identify a function for IL-12 that is tonically produced by type 1 dendritic cells in an MyD88- and CD40-dependent, but foreign PAMP-independent manner.
Collapse
|
208
|
Paracatu LC, Schuettpelz LG. Contribution of Aberrant Toll Like Receptor Signaling to the Pathogenesis of Myelodysplastic Syndromes. Front Immunol 2020; 11:1236. [PMID: 32625214 PMCID: PMC7313547 DOI: 10.3389/fimmu.2020.01236] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Toll like receptors (TLRs) are a family of pattern recognition receptors that play a central role in the innate immune response. These receptors are expressed on a wide variety of immune and non-immune cells, and they help shape the immune response to infection and injury through the recognition of pathogen-associated molecular patterns (PAMPs) as well as endogenous damage-associated molecular patterns (DAMPs). Accumulating evidence suggests that, in addition to regulating mature effector immune cells, TLRs can influence the immune response from the level of the hematopoietic stem cell (HSC). HSCs express TLRs, and exposure to TLR ligands influences the cycling, differentiation, and function of HSCs, with chronic TLR stimulation leading to impairment of normal HSC repopulating activity. Moreover, enhanced TLR expression and signaling is associated with myelodysplastic syndromes (MDS), a heterogenous group of HSC disorders characterized by ineffective hematopoiesis and a high risk of transformation to acute leukemias. In this review, we will discuss the role of TLR signaling in the pathogenesis of MDS, focusing on the known direct and indirect effects of this type of signaling on HSCs, the mechanisms of TLR signaling upregulation in MDS, the changes in TLR expression with disease progression, and the therapeutic implications for modulating TLR signaling in the treatment of MDS.
Collapse
Affiliation(s)
- Luana Chiquetto Paracatu
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Laura G Schuettpelz
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
209
|
Rizzo C, Grasso G, Destro Castaniti GM, Ciccia F, Guggino G. Primary Sjogren Syndrome: Focus on Innate Immune Cells and Inflammation. Vaccines (Basel) 2020; 8:vaccines8020272. [PMID: 32503132 PMCID: PMC7349953 DOI: 10.3390/vaccines8020272] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022] Open
Abstract
Primary Sjogren Syndrome (pSS) is a complex, multifactorial rheumatic disease that mainly targets salivary and lacrimal glands, inducing epithelitis. The cause behind the autoimmunity outbreak in pSS is still elusive; however, it seems related to an aberrant reaction to exogenous triggers such as viruses, combined with individual genetic pre-disposition. For a long time, autoantibodies were considered as the hallmarks of this disease; however, more recently the complex interplay between innate and adaptive immunity as well as the consequent inflammatory process have emerged as the main mechanisms of pSS pathogenesis. The present review will focus on innate cells and on the principal mechanisms of inflammation connected. In the first part, an overview of innate cells involved in pSS pathogenesis is provided, stressing in particular the role of Innate Lymphoid Cells (ILCs). Subsequently we have highlighted the main inflammatory pathways, including intra- and extra-cellular players. A better knowledge of such processes could determine the detection of new therapeutic targets that are a major need for pSS.
Collapse
Affiliation(s)
- Chiara Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Piazza delle Cliniche 2, 90110 Palermo, Italy; (C.R.); (G.G.); (G.M.D.C.)
| | - Giulia Grasso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Piazza delle Cliniche 2, 90110 Palermo, Italy; (C.R.); (G.G.); (G.M.D.C.)
| | - Giulia Maria Destro Castaniti
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Piazza delle Cliniche 2, 90110 Palermo, Italy; (C.R.); (G.G.); (G.M.D.C.)
| | - Francesco Ciccia
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy;
| | - Giuliana Guggino
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Rheumatology Section, University of Palermo, Piazza delle Cliniche 2, 90110 Palermo, Italy; (C.R.); (G.G.); (G.M.D.C.)
- Correspondence: ; Tel.: +39-091-6552260
| |
Collapse
|
210
|
Chuang YC, Tseng JC, Huang LR, Huang CM, Huang CYF, Chuang TH. Adjuvant Effect of Toll-Like Receptor 9 Activation on Cancer Immunotherapy Using Checkpoint Blockade. Front Immunol 2020; 11:1075. [PMID: 32547560 PMCID: PMC7274158 DOI: 10.3389/fimmu.2020.01075] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/04/2020] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy using checkpoint blockade has revolutionized cancer treatment, improving patient survival and quality of life. Nevertheless, the clinical outcomes of such immunotherapy are highly heterogeneous between patients. Depending on the cancer type, the patient response rates to this immunotherapy are limited to 20–30%. Based on the mechanism underlying the antitumor immune response, new therapeutic strategies have been designed with the aim of increasing the effectiveness and specificity of the antitumor immune response elicited by checkpoint blockade agents. The activation of toll-like receptor 9 (TLR9) by its synthetic agonists induces the antitumor response within the innate immunity arm, generating adjuvant effects and priming the adaptive immune response elicited by checkpoint blockade during the effector phase of tumor-cell killing. This review first describes the underlying mechanisms of action and current status of monotherapy using TLR9 agonists and immune checkpoint inhibitors for cancer immunotherapy. The rationale for combining these two agents is discussed, and evidence indicating the current status of such combination therapy as a novel cancer treatment strategy is presented.
Collapse
Affiliation(s)
- Yu-Chen Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Jen-Chih Tseng
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Li-Rung Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Chun-Ming Huang
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
211
|
Gellen B, Thorin-Trescases N, Thorin E, Gand E, Sosner P, Brishoual S, Rigalleau V, Montaigne D, Javaugue V, Pucheu Y, Gatault P, Piguel X, Hadjadj S, Saulnier PJ. Serum tenascin-C is independently associated with increased major adverse cardiovascular events and death in individuals with type 2 diabetes: a French prospective cohort. Diabetologia 2020; 63:915-923. [PMID: 32040670 DOI: 10.1007/s00125-020-05108-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/13/2020] [Indexed: 12/25/2022]
Abstract
AIMS/HYPOTHESIS Tenascin-C (TN-C) is an extracellular matrix glycoprotein highly expressed in inflammatory and cardiovascular (CV) diseases. Serum TN-C has not yet been specifically studied in individuals with type 2 diabetes, a condition associated with chronic low-grade inflammation and increased CV disease risk. In this study, we hypothesised that elevated serum TN-C at enrolment in participants with type 2 diabetes would be associated with increased risk of death and major adverse CV events (MACE) during follow-up. METHODS We used a prospective, monocentric cohort of consecutive type 2 diabetes participants (the SURDIAGENE [SUivi Rénal, DIAbète de type 2 et GENEtique] cohort) with all-cause death as a primary endpoint and MACE (CV death, non-fatal myocardial infarction or stroke) as a secondary endpoint. We used a proportional hazard model after adjustment for traditional risk factors and the relative integrated discrimination improvement (rIDI) to assess the incremental predictive value of TN-C for these risk factors. RESULTS We monitored 1321 individuals (58% men, mean age 64 ± 11 years) for a median of 89 months. During follow-up, 442 individuals died and 497 had MACE. Multivariate Cox analysis showed that serum TN-C concentrations were associated with an increased risk of death (HR per 1 SD: 1.27 [95% CI 1.17, 1.38]; p < 0.0001) and MACE (HR per 1 SD: 1.23 [95% CI 1.13, 1.34]; p < 0.0001). Using TN-C concentrations on top of traditional risk factors, prediction of the risk of all-cause death (rIDI: 8.2%; p = 0.0006) and MACE (rIDI: 6.7%; p = 0.0014) improved significantly, but modestly. CONCLUSIONS/INTERPRETATION In individuals with type 2 diabetes, increased serum TN-C concentrations were independently associated with death and MACE. Therefore, including TN-C as a prognostic biomarker could improve risk stratification in these individuals.
Collapse
Affiliation(s)
- Barnabas Gellen
- ELSAN, Polyclinique de Poitiers, 1 Rue de la Providence, F-86000, Poitiers, France.
| | | | - Eric Thorin
- Research Center, Montreal Heart Institute, Montreal, QC, Canada
- Department of Surgery, Faculty of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Elise Gand
- INSERM, Centre d'Investigation Clinique CIC1402, Université de Poitiers, CHU de Poitiers, Poitiers, France
| | - Philippe Sosner
- INSERM, Centre d'Investigation Clinique CIC1402, Université de Poitiers, CHU de Poitiers, Poitiers, France
- Laboratoire MOVE (EA 6314), Université de Poitiers, Poitiers, France
- Centre Médico-Sportif Mon Stade, Paris, France
| | - Sonia Brishoual
- INSERM, Centre d'Investigation Clinique CIC1402, Université de Poitiers, CHU de Poitiers, Poitiers, France
| | - Vincent Rigalleau
- Endocrinology - Diabetology - Nutrition, CHU de Bordeaux, Hôpital Haut-Lévêque, Pessac, France
| | - David Montaigne
- Department of Clinical Physiology - Echocardiography, CHU Lille, Lille, France
- INSERM U1011, EGID, Institut Pasteur de Lille, University of Lille, Lille, France
| | - Vincent Javaugue
- INSERM, Centre d'Investigation Clinique CIC1402, Université de Poitiers, CHU de Poitiers, Poitiers, France
- Nephrology, CHU de Poitiers, Poitiers, France
| | | | - Philippe Gatault
- Transplantation, Immunology and Inflammation (T2I) - EA4245, CHRU de Tours, Nephrology-Hypertension, Dialysis and Renal Transplantation, FHU SUPORT, Université de Tours, Tours, France
| | - Xavier Piguel
- Endocrinology-Diabetology, CHU de Poitiers, Poitiers, France
| | - Samy Hadjadj
- Transplantation, Immunology and Inflammation (T2I) - EA4245, CHRU de Tours, Nephrology-Hypertension, Dialysis and Renal Transplantation, FHU SUPORT, Université de Tours, Tours, France
- L'Institut du Thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| | - Pierre-Jean Saulnier
- INSERM, Centre d'Investigation Clinique CIC1402, Université de Poitiers, CHU de Poitiers, Poitiers, France
| | | |
Collapse
|
212
|
Wang J, Liu Y, Liu Y, Zhu K, Xie A. The association between TLR3 rs3775290 polymorphism and sporadic Parkinson’s disease in Chinese Han population. Neurosci Lett 2020; 728:135005. [DOI: 10.1016/j.neulet.2020.135005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 04/02/2020] [Accepted: 04/19/2020] [Indexed: 12/22/2022]
|
213
|
Stranik J, Kacerovsky M, Vescicik P, Faist T, Jacobsson B, Musilova I. A rodent model of intra-amniotic inflammation/infection, induced by the administration of inflammatory agent in a gestational sac, associated with preterm delivery: a systematic review. J Matern Fetal Neonatal Med 2020; 35:1592-1600. [PMID: 32349576 DOI: 10.1080/14767058.2020.1757063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: Rodents are the most commonly used animals in the study of spontaneous preterm delivery (PTD). Intra-amniotic inflammation/infection is a frequent and important cause of PTD. Intraperitoneal and intrauterine administrations of inflammatory agents are traditional methods to establish a rodent model of PTD associated with inflammation and infection. The intra-amniotic administration of inflammatory or infectious triggering agents to rodents can be useful to study not only intra-amniotic inflammatory response but also PTD associated with intra-amniotic inflammation/infection.Objective: This systematic review aimed mainly to assess and analyze all described methods of intra-amniotic administration of infectious and/or inflammatory agents to create a rodent model of intra-amniotic inflammation associated with PTD.Methods: A literature search through two electronic databases from their earliest entries to February 2019 was performed. The selection criteria were as follows: (1) rodents as model animals, (2) a model of intra-amniotic inflammation/infection associated with PTD, and (3) intra-amniotic administration of triggering agents. Data extraction included specification of the study (author and year of publication), characteristics of study animals (species, strain, and number of animals), characteristics of intervention (timing and used technique), substance used for induction of intra-amniotic inflammation/infection, and outcome assessment.Results: The search identified a total of 4673 articles, of which 118 were selected for full-text reading, but only 13 studies were included in the review. Intra-amniotic administration was used only in the articles that were published beyond 2004. Two different approaches were identified: (1) open surgery with direct puncture of the amniotic sacs and (2) transabdominal ultrasound-guided puncture of the gestational sacs. Live microorganisms (Ureaplasma parvum), bacterial products (extracellular membrane vesicles), and pathogen-associated (lipopolysaccharide) and damage-associated molecular patterns (high mobility group box-1, S100B, and surfactant protein A) were used to simulate intra-amniotic inflammation/infection. Differences in the effect on intra-amniotic inflammation/infection associated with PTD in the mouse model were identified among triggering agents. Intra-amniotic application of lipopolysaccharide in the rat model caused intra-amniotic inflammation, but it did not lead to PTD.Conclusion: The intra-amniotic administration of the triggering agents can be used to study intra-amniotic inflammatory response and intra-amniotic inflammation/infection in the rodents model.
Collapse
Affiliation(s)
- Jaroslav Stranik
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.,Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Peter Vescicik
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Tomas Faist
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden.,Department of Genetics and Bioinformatics, Domain of Health Data and Digitalisation, Institute of Public Health, Oslo, Norway
| | - Ivana Musilova
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
214
|
Asghar S, Litherland GJ, Lockhart JC, Goodyear CS, Crilly A. Exosomes in intercellular communication and implications for osteoarthritis. Rheumatology (Oxford) 2020; 59:57-68. [PMID: 31628481 DOI: 10.1093/rheumatology/kez462] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/06/2019] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent of the musculoskeletal conditions and represents a significant public health burden. While degeneration of articular cartilage is a key feature, it is now increasingly recognized as a complex condition affecting the whole joint, with synovial inflammation present in a significant proportion of patients. As a secretory tissue, the OA synovium is a rich source of both soluble inflammatory mediators and extracellular vesicles, including exosomes, which have been implicated in cell-cell communication. Exosome cargo has been found to include proteins, lipids and various RNA subtypes such as mRNA and miRNA, potentially capable of regulating gene expression in target cells and tissues. Profiling of exosome cargo and understanding effects on cartilage could elucidate novel regulatory mechanisms within the joint, providing insight for targeted treatment. The aim of this article is to review current literature on exosome biology, highlighting the relevance and application for OA pathogenesis.
Collapse
Affiliation(s)
- Sabha Asghar
- School of Health and Life Sciences, University of the West of Scotland, Paisley Campus, Paisley, UK
| | - Gary J Litherland
- School of Health and Life Sciences, University of the West of Scotland, Paisley Campus, Paisley, UK
| | - John C Lockhart
- School of Health and Life Sciences, University of the West of Scotland, Paisley Campus, Paisley, UK
| | - Carl S Goodyear
- Institute of Infection, Immunity and Inflammation, GBRC, University Place, University of Glasgow, Glasgow, UK
| | - Anne Crilly
- School of Health and Life Sciences, University of the West of Scotland, Paisley Campus, Paisley, UK
| |
Collapse
|
215
|
Vasanthakumar N, Bhakta-Guha D, Guha G, Arunachalam J. Friend turned foe: A curious case of disrupted endosymbiotic homeostasis promoting the Warburg effect in sepsis. Med Hypotheses 2020; 141:109702. [PMID: 32289643 DOI: 10.1016/j.mehy.2020.109702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 03/30/2020] [Indexed: 10/24/2022]
Abstract
Sepsis is a grievous health concern with limited understanding of its precise etiology. Although studies on sepsis have implicated the Warburg effect (mitigation of mitochondrial oxidative phosphorylation, as evident from aerobic glycolysis), we propose that an evolutionary perspective might further unravel its etiology. The endosymbiotic theory suggests that evolution of a eukaryotic cell is a consequence of the fruitful association between an archaea (Asgard) and an alphaproteobacterium (Rickettsia). We hypothesize that, during pathological conditions like sepsis, such endosymbiotic homeostasis between the two systems is perturbed. We underscore the fact (supported by in silico homology analyses) that during sepsis, the Asgard component of a cell is promoted to trigger aerobic glycolysis as well as the innate immune response (spearheaded by the TLR pathway), while suppressing the Rickettsia counterpart, thereby promoting the Warburg effect. It might be this discord between the two endosymbiotic partners (Asgard and Rickettsia-derived cellular components) that promotes sepsis.
Collapse
Affiliation(s)
- Natesan Vasanthakumar
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu 613401, India.
| | - Dipita Bhakta-Guha
- Cellular Dyshomeostasis Laboratory (CDHL), School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu 613401, India
| | - Gunjan Guha
- Cellular Dyshomeostasis Laboratory (CDHL), School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu 613401, India
| | - Jothi Arunachalam
- Department of Bioinformatics, School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu 613401, India
| |
Collapse
|
216
|
Robertson SA, Hutchinson MR, Rice KC, Chin PY, Moldenhauer LM, Stark MJ, Olson DM, Keelan JA. Targeting Toll-like receptor-4 to tackle preterm birth and fetal inflammatory injury. Clin Transl Immunology 2020; 9:e1121. [PMID: 32313651 PMCID: PMC7156293 DOI: 10.1002/cti2.1121] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 12/30/2022] Open
Abstract
Every year, 15 million pregnancies end prematurely, resulting in more than 1 million infant deaths and long-term health consequences for many children. The physiological processes of labour and birth involve essential roles for immune cells and pro-inflammatory cytokines in gestational tissues. There is compelling evidence that the mechanisms underlying spontaneous preterm birth are initiated when a premature and excessive inflammatory response is triggered by infection or other causes. Exposure to pro-inflammatory mediators is emerging as a major factor in the 'fetal inflammatory response syndrome' that often accompanies preterm birth, where unscheduled effects in fetal tissues interfere with normal development and predispose to neonatal morbidity. Toll-like receptors (TLRs) are critical upstream gatekeepers of inflammatory activation. TLR4 is prominently involved through its ability to sense and integrate signals from a range of microbial and endogenous triggers to provoke and perpetuate inflammation. Preclinical studies have identified TLR4 as an attractive pharmacological target to promote uterine quiescence and protect the fetus from inflammatory injury. Novel small-molecule inhibitors of TLR4 signalling, specifically the non-opioid receptor antagonists (+)-naloxone and (+)-naltrexone, are proving highly effective in animal models for preventing preterm birth induced by bacterial mimetic LPS, heat-killed Escherichia coli, or the TLR4-dependent pro-inflammatory lipid, platelet-activating factor (PAF). Here, we summarise the rationale for targeting TLR4 as a master regulator of inflammation in fetal and gestational tissues, and the potential utility of TLR4 antagonists as candidates for preventative and therapeutic application in preterm delivery and fetal inflammatory injury.
Collapse
Affiliation(s)
- Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School University of Adelaide Adelaide SA Australia
| | - Mark R Hutchinson
- Robinson Research Institute and Adelaide Medical School University of Adelaide Adelaide SA Australia.,ARC Centre for Nanoscale Biophotonics and Adelaide Medical School University of Adelaide Adelaide SA Australia
| | - Kenner C Rice
- Drug Design and Synthesis Section National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism National Institutes of Health Rockville MD USA
| | - Peck-Yin Chin
- Robinson Research Institute and Adelaide Medical School University of Adelaide Adelaide SA Australia
| | - Lachlan M Moldenhauer
- Robinson Research Institute and Adelaide Medical School University of Adelaide Adelaide SA Australia
| | - Michael J Stark
- Robinson Research Institute and Adelaide Medical School University of Adelaide Adelaide SA Australia
| | - David M Olson
- Department of Obstetrics and Gynecology Department of Physiology and Pediatrics 220 HMRC University of Alberta Edmonton AB Canada
| | - Jeffrey A Keelan
- Division of Obstetrics & Gynaecology University of Western Australia Perth WA Australia
| |
Collapse
|
217
|
Chen C, Li J, Zhang W, Shah SWA, Ishfaq M. Mycoplasma gallisepticum triggers immune damage in the chicken thymus by activating the TLR-2/MyD88/NF-κB signaling pathway and NLRP3 inflammasome. Vet Res 2020; 51:52. [PMID: 32276652 PMCID: PMC7149927 DOI: 10.1186/s13567-020-00777-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/29/2020] [Indexed: 02/06/2023] Open
Abstract
Previous studies reported that Mycoplasma gallisepticum (MG) causes immune dysregulation in chickens. However, the underlying mechanisms of immune dysregulation in chickens are still unclear. The thymus is a primary lymphoid organ where the proliferation, differentiation and selection of T-lymphocytes occur, whereas T-lymphocytes play a crucial role in innate immune responses. To evaluate the effects of MG-infection on chicken thymus, White Leghorn chickens were divided into (1) control group and (2) MG-infection group. ATPase activities were detected by commercial kits. The hallmarks of inflammation, autophagy and energy metabolism were examined in chicken thymus tissues by histopathology, transmission electron microscopy, immunofluorescence microscopy, RT-PCR and western blotting. Immunofluorescence examination revealed that the number of CD8+ lymphocytes has significantly reduced in MG-infection group. In addition, morphological analysis revealed that MG induced inflammatory cells infiltration. The mitochondria were swollen and chromatin material was condensed in MG-infection group. The mRNA and protein expression results showed that MG-infection triggered the nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain containing 3 (NLRP3) inflammasome through TLR-2/MyD88/NF-κB signaling pathway. Meanwhile, the expressions of autophagy-related genes were reduced both at mRNA and protein level in MG-infection group. While, ATPase activities and the expression of energy metabolism-related genes were reduced in the thymus of MG-infected chickens. These results showed that MG-infection triggered inflammatory response through TLR-2/MyD88/NF-κB signaling pathway, activated NLRP3 inflammasome, reduced the level of autophagy and impaired energy metabolism, which then lead to tissue damage in chicken thymus. The data provide new insights in MG-infection-mediated immune damage and provide possible therapeutic targets for future targeted therapy.
Collapse
Affiliation(s)
- Chunli Chen
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, 150030, China
| | - Jichang Li
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, 150030, China
| | - Wei Zhang
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, 150030, China
| | - Syed Waqas Ali Shah
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Muhammad Ishfaq
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin, 150030, China.
| |
Collapse
|
218
|
Keshavarzian A, Engen P, Bonvegna S, Cilia R. The gut microbiome in Parkinson's disease: A culprit or a bystander? PROGRESS IN BRAIN RESEARCH 2020; 252:357-450. [PMID: 32247371 DOI: 10.1016/bs.pbr.2020.01.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In recent years, large-scale metagenomics projects such as the Human Microbiome Project placed the gut microbiota under the spotlight of research on its role in health and in the pathogenesis several diseases, as it can be a target for novel therapeutical approaches. The emerging concept of a microbiota modulation of the gut-brain axis in the pathogenesis of neurodegenerative disorders has been explored in several studies in animal models, as well as in human subjects. Particularly, research on changes in the composition of gut microbiota as a potential trigger for alpha-synuclein (α-syn) pathology in Parkinson's disease (PD) has gained increasing interest. In the present review, we first provide the basis to the understanding of the role of gut microbiota in healthy subjects and the molecular basis of the gut-brain interaction, focusing on metabolic and neuroinflammatory factors that could trigger the alpha-synuclein conformational changes and aggregation. Then, we critically explored preclinical and clinical studies reporting on the changes in gut microbiota in PD, as compared to healthy subjects. Furthermore, we examined the relationship between the gut microbiota and PD clinical features, discussing data consistently reported across studies, as well as the potential sources of inconsistencies. As a further step toward understanding the effects of gut microbiota on PD, we discussed the relationship between dysbiosis and response to dopamine replacement therapy, focusing on Levodopa metabolism. We conclude that further studies are needed to determine whether the gut microbiota changes observed so far in PD patients is the cause or, instead, it is merely a consequence of lifestyle changes associated with the disease. Regardless, studies so far strongly suggest that changes in microbiota appears to be impactful in pathogenesis of neuroinflammation. Thus, dysbiotic microbiota in PD could influence the disease course and response to medication, especially Levodopa. Future research will assess the impact of microbiota-directed therapeutic intervention in PD patients.
Collapse
Affiliation(s)
- Ali Keshavarzian
- Department of Internal Medicine, Division of Digestive Disease and Nutrition, Rush University Medical Center, Chicago, IL, United States
| | - Phillip Engen
- Department of Internal Medicine, Division of Digestive Disease and Nutrition, Rush University Medical Center, Chicago, IL, United States
| | | | - Roberto Cilia
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Movement Disorders Unit, Milan, Italy.
| |
Collapse
|
219
|
Abstract
Cutaneous adverse drug reactions are unpredictable and include various different skin conditions of varying degrees of severity. The most concerning are usually referred to as severe cutaneous adverse reactions (SCARs) and include acute generalized exanthematous pustulosis (AGEP), drug reaction with eosinophilia and systemic symptoms (DRESS), also known as drug-induced hypersensitivity syndrome (DiHS) or hypersensitivity syndrome (HSS), Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN). All are delayed type IV hypersensitivity reactions in which a T-cell-mediated drug-specific immune response is responsible for causing the disease. Nonetheless, specific T-cell subpopulations develop in response to certain environmental conditions and produce cytokines that orchestrate the various phenotypes. Cytotoxic T lymphocytes (CTLs), T-helper type 1 (Th1), Th2, Th17, and regulatory T cells (Treg), among other T-cell subpopulations, participate in the development of SCAR phenotypes. Cell subpopulations belonging to the innate immune system, comprising natural killer cells, innate lymphoid cells, monocytes, macrophages and dendritic cells, can also participate in shaping specific immune responses in various clinical conditions. Additionally, tissue-resident cells, including keratinocytes, can contribute to epidermal damage by secreting chemokines that attract pro-inflammatory immunocytes. The final phenotypes in each clinical entity result from the complex interactions between a variety of cell lineages, their products, soluble mediators and genetic and environmental factors. Although the pathophysiology of these reactions is not fully understood, intensive research in recent years has led to major progress in our understanding of the contribution of certain cell types and soluble mediators to the variability of SCAR phenotypes.
Collapse
Affiliation(s)
- Teresa Bellón
- La Paz Hospital Health Research Institute-IdiPAZ, Pº Castellana 261, 28046, Madrid, Spain.
| |
Collapse
|
220
|
Bhutta MF, Lambie J, Hobson L, Williams D, Tyrer HE, Nicholson G, Brown SDM, Brown H, Piccinelli C, Devailly G, Ramsden J, Cheeseman MT. Transcript Analysis Reveals a Hypoxic Inflammatory Environment in Human Chronic Otitis Media With Effusion. Front Genet 2020; 10:1327. [PMID: 32153623 PMCID: PMC7047850 DOI: 10.3389/fgene.2019.01327] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic otitis media with effusion (COME) is the most common cause of childhood hearing loss in the developed world. Underlying pathophysiology is not well understood, and in particular the factors that lead to the transition from acute to chronic inflammation. Here we present the first genome-wide transcript analysis of white blood cells in the effusion of children with COME. Analysis of microarray data for enriched pathways reveals upregulation of hypoxia pathways, which is confirmed using real-time PCR and determining VEGF protein titres. Other pathways upregulated in both mucoid and serous effusions include Toll-like receptor signaling, complement, and RANK-RANKL. Cytology reveals neutrophils and macrophages predominated in both serous and mucoid effusions, however, serous samples had higher lymphocyte and eosinophil differential counts, while mucoid samples had higher neutrophil differential counts. Transcript analysis indicates serous fluids have CD4+ and CD8+ T-lymphocyte, and NK cell signatures. Overall, our findings suggest that inflammation and hypoxia pathways are important in the pathology of COME, and targets for potential therapeutic intervention, and that mucoid and serous COME may represent different immunological responses.
Collapse
Affiliation(s)
- Mahmood F Bhutta
- Department of ENT, Brighton & Sussex University Hospitals NHS Trust, Brighton, United Kingdom
| | - Jane Lambie
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Lindsey Hobson
- Department of ENT, Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Debbie Williams
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, United Kingdom
| | - Hayley E Tyrer
- Faculty of Health and Wellbeing, University of Lancashire, Preston, United Kingdom
| | - George Nicholson
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Steve D M Brown
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell, United Kingdom
| | - Helen Brown
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Chiara Piccinelli
- The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| | | | - James Ramsden
- Department of ENT, Oxford University Hospitals NHS Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Michael T Cheeseman
- The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
221
|
Wahid HH, Chin PY, Sharkey DJ, Diener KR, Hutchinson MR, Rice KC, Moldenhauer LM, Robertson SA. Toll-Like Receptor-4 Antagonist (+)-Naltrexone Protects Against Carbamyl-Platelet Activating Factor (cPAF)-Induced Preterm Labor in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1030-1045. [PMID: 32084361 DOI: 10.1016/j.ajpath.2020.01.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/24/2019] [Accepted: 01/17/2020] [Indexed: 02/06/2023]
Abstract
Spontaneous preterm labor is frequently caused by an inflammatory response in the gestational tissues elicited by either infectious or sterile agents. In sterile preterm labor, the key regulators of inflammation are not identified, but platelet-activating factor (PAF) is implicated as a potential rate-limiting effector agent. Since Toll-like receptor (TLR)-4 can amplify PAF signaling, we evaluated whether TLR4 contributes to inflammation and fetal loss in a mouse model of PAF-induced sterile preterm labor, and whether a small-molecule TLR4 inhibitor, (+)-naltrexone, can mitigate adverse PAF-induced effects. The administration of carbamyl (c)-PAF caused preterm labor and fetal loss in wild-type mice but not in TLR4-deficient mice. Treatment with (+)-naltrexone prevented preterm delivery and alleviated fetal demise in utero elicited after cPAF administered by i.p. or intrauterine routes. Pups born after cPAF and (+)-naltrexone treatment exhibited comparable rates of postnatal survival and growth to carrier-treated controls. (+)-Naltrexone suppressed the cPAF-induced expression of inflammatory cytokine genes Il1b, Il6, and Il10 in the decidua; Il6, Il12b, and Il10 in the myometrium; and Il1b and Il6 in the placenta. These data demonstrate that the TLR4 antagonist (+)-naltrexone inhibits the inflammatory cascade induced by cPAF, preventing preterm birth and perinatal death. The inhibition of TLR4 signaling warrants further investigation as a candidate strategy for fetal protection and delay of preterm birth elicited by sterile stimuli.
Collapse
Affiliation(s)
- Hanan H Wahid
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Peck Yin Chin
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - David J Sharkey
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Kerrilyn R Diener
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
| | - Mark R Hutchinson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, South Australia, Australia
| | - Kenner C Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland
| | - Lachlan M Moldenhauer
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
222
|
Hughes H, Ashwood P. Overlapping evidence of innate immune dysfunction in psychotic and affective disorders. Brain Behav Immun Health 2020; 2:100038. [PMID: 34589829 PMCID: PMC8474635 DOI: 10.1016/j.bbih.2020.100038] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/17/2020] [Accepted: 01/24/2020] [Indexed: 12/17/2022] Open
Abstract
Disturbances of the immune system and immune responses after activation are a common finding in neuropsychiatric disorders. Psychotic and affective disorders such as major depressive disorder (MDD), schizophrenia (SCZ) and bipolar disorder (BD) also share high rates of comorbidity with inflammatory and metabolic disorders. Evidence of elevated circulating inflammatory cytokines, altered numbers and function of immune cells, and evidence of neuroinflammation including activation of microglia in the brain have been found in patients with SCZ, BD and MDD. Often these findings correlate to psychological state at the time of measurement. However, significant variation exists across these studies in many aspects, creating challenges in identifying a specific signature of immune dysfunction in these disorders. Innate immune dysfunction, and alterations in monocytes, the critical sentinel cells of the innate immune system, have been seen repeatedly in all three of these disorders, with frequent overlap in findings. In this review, dysfunction specific to the innate arm of the immune system is compared for overlapping evidence across three major psychotic and affective disorders.
Collapse
Affiliation(s)
- H.K. Hughes
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- MIND Institute, University of California Davis, Davis, CA, USA
| | - P. Ashwood
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| |
Collapse
|
223
|
Kiripolsky J, Romano RA, Kasperek EM, Yu G, Kramer JM. Activation of Myd88-Dependent TLRs Mediates Local and Systemic Inflammation in a Mouse Model of Primary Sjögren's Syndrome. Front Immunol 2020; 10:2963. [PMID: 31993047 PMCID: PMC6964703 DOI: 10.3389/fimmu.2019.02963] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/03/2019] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptors (TLRs) are important mediators of chronic inflammation in numerous autoimmune diseases, although the role of these receptors in primary Sjögren's syndrome (pSS) remains incompletely understood. Previous studies in our laboratory established Myd88 as a crucial mediator of pSS, although the disease-relevant ligands and the upstream signaling events that culminate in Myd88 activation have yet to be established. The objective of this study was to identify specific Myd88-dependent TLR-related pathways that are dysregulated both locally and systemically in a mouse model of pSS [NOD.B10Sn-H2b/J (NOD.B10)]. We performed RNA-sequencing on spleens derived from NOD.B10 mice. We then harvested salivary tissue and spleens from Myd88-sufficient and deficient C57BL/10 (BL/10) and NOD.B10 mice and performed flow cytometry to determine expression of Myd88-dependent TLRs. We cultured splenocytes with TLR2 and TLR4 agonists and measured production of inflammatory mediators by ELISA. Next, we evaluated spontaneous and TLR4-mediated inflammatory cytokine secretion in NOD.B10 salivary tissue. Finally, we assessed spontaneous Myd88-dependent cytokine secretion by NOD.B10 salivary cells. We identified dysregulation of numerous TLR-related networks in pSS splenocytes, particularly those employed by TLR2 and TLR4. We found upregulation of TLRs in both the splenic and salivary tissue from pSS mice. In NOD.B10 splenic tissue, robust expression of B cell TLR1 and TLR2 required Myd88. Splenocytes from NOD.B10 mice were hyper-responsive to TLR2 ligation and the endogenous molecule decorin modulated inflammation via TLR4. Finally, we observed spontaneous secretion of numerous inflammatory cytokines and this was enhanced following TLR4 ligation in female NOD.B10 salivary tissue as compared to males. The spontaneous production of salivary IL-6, MCP-1 and TNFα required Myd88 in pSS salivary tissue. Thus, our data demonstrate that Myd88-dependent TLR pathways contribute to the inflammatory landscape in pSS, and inhibition of such will likely have therapeutic utility.
Collapse
Affiliation(s)
- Jeremy Kiripolsky
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States
| | - Rose-Anne Romano
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States
| | - Eileen M Kasperek
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States
| | - Guan Yu
- Department of Biostatistics, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY, United States
| | - Jill M Kramer
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States.,Department of Oral Diagnostic Sciences, School of Dental Medicine, State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
224
|
Hernandez H, Roberts AL, McDowell CM. Nuclear factor-kappa beta signaling is required for transforming growth factor Beta-2 induced ocular hypertension. Exp Eye Res 2020; 191:107920. [PMID: 31923415 DOI: 10.1016/j.exer.2020.107920] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/18/2019] [Accepted: 01/05/2020] [Indexed: 01/22/2023]
Abstract
A major risk for the development of primary open-angle glaucoma (POAG) is elevated intraocular pressure (IOP). Elevated IOP is caused by increased outflow resistance due in part to excessive extracellular matrix (ECM) deposition in the trabecular meshwork (TM). The role of transforming growth factor beta 2 (TGFβ2) in inducing ECM production is well understood. Recent studies suggest that toll-like receptor 4 (TLR4) plays an important role in fibrogenesis. We have previously described a crosstalk between TGFβ2 and TLR4 in the development of ocular hypertension and glaucomatous TM damage. Nuclear factor-kappa beta (NF-κB) is critical for TLR4 signaling. To determine the transactivation of NF-κB, TM cells were stimulated with cellular fibronectin containing the EDA isoform (cFN-EDA), TGFβ2, or lipopolysaccharide (LPS) in combination with a selective TLR4 inhibitor. cFN-EDA, TGFβ2, and LPS all induced transactivation of NF-κB and inhibition of TLR4 blocked the effect of each treatment paradigm. To evaluate the role of NF-κB in IOP regulation, we utilized our inducible mouse model of ocular hypertension by injection of Ad5.TGFβ2 in mice harboring a mutation in NF-κB and wild-type controls. IOP was measured over time and eyes accessed by immunohistochemistry for the ECM protein FN and the specific FN-EDA isoform. Ad5.TGFβ2 induced ocular hypertension and expression of FN and FN-EDA in wild-type mice, but mutation in NF-κB blocked the effect. These data suggest that NF-κB is necessary for TGFβ2-induced ECM production and ocular hypertension and the transactivation of NF-κB is dependent on both TGFβ2 and TLR4.
Collapse
Affiliation(s)
- Humberto Hernandez
- North Texas Eye Research Institute, Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Amanda L Roberts
- North Texas Eye Research Institute, Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Colleen M McDowell
- University of Wisconsin-Madison, Department of Ophthalmology and Visual Sciences, Madison, WI, 53706, USA.
| |
Collapse
|
225
|
Cagliani J, Yang WL, Brenner M, Wang P. Deoxyribonuclease Reduces Tissue Injury and Improves Survival After Hemorrhagic Shock. J Surg Res 2020; 249:104-113. [PMID: 31926397 DOI: 10.1016/j.jss.2019.11.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/16/2019] [Accepted: 11/23/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Hemorrhagic shock (HS) caused by rapid loss of a large amount of blood is the leading cause of early death after severe injury. When cells are damaged during HS, many intracellular components including DNA are released into the circulation and function as endogenous damage-associated molecular patterns (DAMPs) that can trigger excessive inflammatory response and subsequently multiple organ dysfunction. We hypothesized that the administration of deoxyribonuclease I (DNase I) could reduce cell-free DNA and attenuate tissue damage in HS. METHODS Eight-week-old male C57BL/6 mice underwent HS by controlled bleeding from the femoral artery for 90 min, followed by resuscitation with Ringer's lactate solution (vehicle) or DNase I (10 mg/kg BW). RESULTS At 20 h after HS, serum levels of cell-free DNA were increased by 7.6-fold in the vehicle-treated HS mice compared with sham, while DNase I reduced its levels by 47% compared with the vehicle group. Serum levels of tissue injury markers (lactate dehydrogenase, aspartate aminotransferase, and alanine aminotransferase) and proinflammatory cytokine interleukin 6 were significantly reduced in the DNase I-treated mice. In the lungs, messenger RNA levels of proinflammatory cytokines (interleukin 6 and interleukin 1 β), chemoattractant macrophage inflammatory protein - 2, and myeloperoxidase activity were significantly decreased in HS mice after DNase I. Finally, DNase I significantly improved the 10-day survival rate in HS mice. CONCLUSIONS Administration of DNase I attenuates tissue damage and systemic and lung inflammation, leading to improvement of survival in HS mice. Thus, DNase I may potentially serve as an adjunct therapy for managing patients with HS.
Collapse
Affiliation(s)
- Joaquin Cagliani
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, New York; Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York; Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York
| | - Weng-Lang Yang
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, New York; Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York; Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York
| | - Max Brenner
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, New York; Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York
| | - Ping Wang
- Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, New York; Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York.
| |
Collapse
|
226
|
González-Quilen C, Rodríguez-Gallego E, Beltrán-Debón R, Pinent M, Ardévol A, Blay MT, Terra X. Health-Promoting Properties of Proanthocyanidins for Intestinal Dysfunction. Nutrients 2020; 12:E130. [PMID: 31906505 PMCID: PMC7019584 DOI: 10.3390/nu12010130] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/20/2019] [Accepted: 12/31/2019] [Indexed: 12/22/2022] Open
Abstract
The intestinal barrier is constantly exposed to potentially harmful environmental factors, including food components and bacterial endotoxins. When intestinal barrier function and immune homeostasis are compromised (intestinal dysfunction), inflammatory conditions may develop and impact overall health. Evidence from experimental animal and cell culture studies suggests that exposure of intestinal mucosa to proanthocyanidin (PAC)-rich plant products, such as grape seeds, may contribute to maintaining the barrier function and to ameliorating the pathological inflammation present in diet-induced obesity and inflammatory bowel disease. In this review, we aim to update the current knowledge on the bioactivity of PACs in experimental models of intestinal dysfunction and in humans, and to provide insights into the underlying biochemical and molecular mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | - M Teresa Blay
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (C.G.-Q.); (E.R.-G.); (R.B.-D.); (M.P.); (A.A.); (X.T.)
| | | |
Collapse
|
227
|
Viswanathan VK, Shetty AP, Rajasekaran S. Modic changes - An evidence-based, narrative review on its patho-physiology, clinical significance and role in chronic low back pain. J Clin Orthop Trauma 2020; 11:761-769. [PMID: 32879563 PMCID: PMC7452231 DOI: 10.1016/j.jcot.2020.06.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/12/2020] [Accepted: 06/15/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Lumbar degenerative spinal ailments are the most important causes for chronic low back pain. Modic changes (MC) are vertebral bone marrow signal intensity changes seen on MRI, commonly in association with degenerative disc disease (DDD). Despite being widely studied, majority of issues concerning MC are still controversial. The current narrative, evidence-based review comprehensively discusses the various aspects related to MC. LITERATURE SEARCH An elaborate search was made using keywords "Modic changes", "lumbar Modic changes", "Modic changes in lumbar spine", and "vertebral Endplate Spinal Changes", on pubmed and google (scholar.google.com) databases on the 3rd of March 2020. We identified crucial questions regarding Modic changes and included relevant articles pertaining to these topics for this narrative review. RESULTS The initial search using the keywords "Modic changes", "lumbar Modic changes", "Modic changes in lumbar spine", and "vertebral Endplate Spinal Changes" on pubmed yielded a total of 568, 412, 394 and 216 articles on "pubmed" database, respectively. A similar search using the aforementioned keywords yielded a total of 3650, 3548, 3726 and 21570 articles on "google scholar" database. The initial screening involved exclusion of duplicate articles, articles unrelated to MC, animal or other non-clinical studies, and articles in non-English literature based on abstracts or the titles of articles. This initial screening resulted in the identification of 405 articles. Full manuscripts were obtained for all these selected articles and thoroughly scrutinised at the second stage of article selection. All articles not concerning Modic changes, not pertaining to concerned questions, articles concerning other degenerative phenomena, articles discussing cervical or thoracic MC, case reports or animal studies, articles in non-English language and duplicate articles were excluded. Review articles, randomised controlled trials and level 1 studies were given preference. Overall, 69 articles were included in this review. CONCLUSION Modic change (MC) is a dynamic phenomenon and its true etiology is still not definitely known. Disc/end plate injury, occult discitis and autoimmune reactions seem to trigger an inflammatory cascade, which leads to their development. Male sex, older age, diabetes mellitus, genetic factors, smoking, obesity, spinal deformities, higher occupational loads and DDD are known risk factors. There is no conclusive evidence on the causative role of MC in chronic low back pain (LBP) or any influence on the long term outcome in patients with LBP or lumbar disc herniations (LDH). Patients with MC have been reported to have less satisfactory outcome following conservative treatment or discectomy, although the evidence is still unclear.
Collapse
Affiliation(s)
| | - Ajoy Prasad Shetty
- Department of Spine Surgery, Ganga Medical Center and Hospitals, Coimbatore, India,Corresponding author.
| | - S. Rajasekaran
- Department of Spine Surgery, Ganga Medical Center and Hospitals, Coimbatore, India,Department of Orthopedics, Ganga Medical Center and Hospitals, Coimbatore, India
| |
Collapse
|
228
|
Suzuki JI, Miki S, Ushijima M, Kodera Y. Regulation of immune response by S-1-propenylcysteine through autophagy-mediated protein degradation. Exp Ther Med 2019; 19:1570-1573. [PMID: 32010341 PMCID: PMC6966193 DOI: 10.3892/etm.2019.8392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/06/2019] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a key event in cellular recycling processes due to its involvement in the intracellular degradation of proteins. It has been demonstrated that S−1-propenylcysteine (S1PC), a characteristic sulfur compound in aged garlic extract, induces the activation of autophagy. S1PC degrades the adaptor protein myeloid differentiation response protein 88 (MyD88) of downstream of Toll-like receptor (TLR) by activating autophagy in vitro and in vivo. The degradation of MyD88 inhibits the TLR signaling pathway, including the phosphorylation of interleukin 1 receptor associated kinase 4 (IRAK4) and nuclear factor (NF)-κB p65 in vitro, and eventually leads to the inhibition of interleukin (IL)-6 production in vitro and C-C motif chemokine ligand 2 (Ccl2) mRNA expression in vivo. S1PC also increases the level of intestinal immunoglobulin A (IgA) and the number of IgA-producing cells in Peyer's patches in vivo. In addition, S1PC triggers the mRNA expression of X-box binding protein 1 (Xbp1), an inducer of IgA-producing cell differentiation via the phosphorylation of extracellular signal-regulated kinase (ERK)1/2 and the degradation of paired box protein 5 (Pax5), a suppressor of Xbp1 mRNA expression. The present review summarizes the mechanisms through which the activation of autophagy by S1PC modulates the immune response.
Collapse
Affiliation(s)
- Jun-Ichiro Suzuki
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| | - Satomi Miki
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| | - Mitsuyasu Ushijima
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| | - Yukihiro Kodera
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| |
Collapse
|
229
|
Miki S, Suzuki JI, Kunimura K, Morihara N. Mechanisms underlying the attenuation of chronic inflammatory diseases by aged garlic extract: Involvement of the activation of AMP-activated protein kinase. Exp Ther Med 2019; 19:1462-1467. [PMID: 32010323 PMCID: PMC6966139 DOI: 10.3892/etm.2019.8372] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/09/2019] [Indexed: 02/06/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is an ubiquitously expressed serine/threonine kinase and an important regulator of energy metabolism. The decreased activity of AMPK induced by low-grade chronic inflammation has been implicated in several diseases, including type 2 diabetes and atherosclerosis. However, the activation of AMPK by natural and synthetic products can ameliorate these diseases through the inhibition of inflammation. For example, aged garlic extract (AGE) has been shown to enhance the phosphorylation of Thr172 of the α-subunit of AMPK in several tissues of disease model animals. In addition, AGE has been reported to suppress the progression of atherosclerotic plaque formation in an animal model of atherosclerosis. Moreover, AGE has been found to decrease the level of plasma glycated albumin and to improve hyperglycemia in an animal model of type 2 diabetes. These inhibitory effects of AGE are induced by the suppression of the inflammatory response. In the present review, we discuss the mechanisms through which AGE activates AMPK, as well as the mechanisms through which the activation of AMPK by AGE modulates the inflammatory response in disease models.
Collapse
Affiliation(s)
- Satomi Miki
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Akitakata-shi, Hiroshima 739-1195, Japan
| | - Jun-Ichiro Suzuki
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Akitakata-shi, Hiroshima 739-1195, Japan
| | - Kayo Kunimura
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Akitakata-shi, Hiroshima 739-1195, Japan
| | - Naoaki Morihara
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Akitakata-shi, Hiroshima 739-1195, Japan.,Research and Development, Wakunaga of America Co., Ltd., Mission Viejo, CA 92691, USA
| |
Collapse
|
230
|
Sharma TP, Curry S, McDowell CM. Effects of Toll-Like Receptor 4 Inhibition on Transforming Growth Factor-β2 Signaling in the Human Trabecular Meshwork. J Ocul Pharmacol Ther 2019; 36:170-178. [PMID: 31834824 DOI: 10.1089/jop.2019.0076] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose: Transforming growth factor-β2 (TGFβ2) and Toll-like receptor 4 (TLR4) crosstalk have been implicated in extracellular matrix regulation in the trabecular meshwork (TM) and ocular hypertension in mice. We investigated TLR4 expression in normal and glaucomatous human trabecular meshwork (HTM) sections and utilized a human perfusion organ culture model to determine TGFβ2-TLR4 signaling crosstalk in glaucoma. Methods: Expression of TLR4 was determined in TM of normal and glaucomatous human eyes. Anterior segments of paired human eyes were perfused at a constant flow rate (2.5 μL/min) for 4 days to acquire stable baseline intraocular pressures (IOPs). We treated paired eyes with two different treatment paradigms: (1) TGFβ2 in one eye and vehicle control in the paired eye, (2) TGFβ2 in one eye and TGFβ2 + TLR4 inhibitor TAK-242 in the paired eye. Perfusate and TM tissue were collected and analyzed for fibronectin (FN) and collagen IV (COLIV) expression. Results: We observed increased TLR4 expression in glaucomatous HTM sections compared to normal (age-matched) (P < 0.05). Significant elevation of IOP was detected in 47% of TGFβ2-treated anterior segments (P < 0.01) compared to control, and in TGFβ2 treated compared with co-treatment with TGFβ2 + TLR4 inhibitor (P < 0.0001). An increase in FN and COLIV expression was observed after TGFβ2 treatment, and inhibition of TLR4 signaling decreased TGFβ2-induced FN and COLIV expression in perfusate (P < 0.05). Conclusions: These studies identify TGFβ2-TLR4 crosstalk as a novel pathway in glaucoma. They provide a potential new target to lower IOP and explore glaucoma pathogenesis.
Collapse
Affiliation(s)
- Tasneem P Sharma
- Department of Pharmacology and Neuroscience, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas
| | - Stacy Curry
- Department of Pharmacology and Neuroscience, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, Texas
| | - Colleen M McDowell
- Department of Ophthalmology and Visual Sciences, McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
231
|
Perčić I, Urošević I, Stokić E, Tomić Naglić D, Milošević I. Febrile neutropenia induces changes in insulin sensitivity similar to obesity. Acta Clin Belg 2019; 74:393-398. [PMID: 30332920 DOI: 10.1080/17843286.2018.1534577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Background/aim: To determine insulin sensitivity before chemotherapy and during febrile neutropenia in patients with acute leukemia and to assess its effect on the number of documented infections, the severity of infection and the outcome of the first hospitalization. To compare insulin sensitivity in the study group to a group of patient with obesity. Materials and methods: The study group consisted of 30 (37% of the total number) patients with newly diagnosed acute leukemia. Testing of insulin sensitivity was done before chemotherapy and during febrile neutropenia. Parameters were compared to a group of 30 age, and sex matched patients with obesity. Results: Insulin sensitivity was normal before chemotherapy. Obese patients were characterized by insulin resistance. Febrile neutropenia led to the development of insulin resistance (t = -2.43, p = 0.021). The level of insulin resistance was in positive correlation with fibrinogen (r = 0.59, p < 0.05). Patients with a documented site of infection had higher fasting insulin and an insulin resistance before chemotherapy (t = -2.38, p = 0.024). Insulin sensitivity did not influence outcome of the first hospitalization. Conclusion: Patients with acute leukemia in febrile neutropenia developed changes in insulin sensitivity similar to those seen in obesity. Insulin resistance was present in patients with a documented site of infection, and it worsened with the extent of inflammation. The outcome of the first hospitalization was not affected.
Collapse
Affiliation(s)
- Ivanka Perčić
- Clinic of Hematology, Clinical Center Vojvodina, Medical Faculty, University of Novi Sad, Novi Sad, Serbia
| | - Ivana Urošević
- Clinic of Hematology, Clinical Center Vojvodina, Medical Faculty, University of Novi Sad, Novi Sad, Serbia
| | - Edita Stokić
- Clinic of Diabetes, Endocrinology and Metabolic Disorders, Clinical Center Vojvodina, Medical Faculty, University of Novi Sad, Novi Sad, Serbia
| | - Dragana Tomić Naglić
- Clinic of Diabetes, Endocrinology and Metabolic Disorders, Clinical Center Vojvodina, Medical Faculty, University of Novi Sad, Novi Sad, Serbia
| | - Ivana Milošević
- Clinic of Hematology, Clinical Center Vojvodina, Medical Faculty, University of Novi Sad, Novi Sad, Serbia
| |
Collapse
|
232
|
Lee B, Song YS, Rhodes C, Goh TS, Roh JS, Jeong H, Park J, Lee HN, Lee SG, Kim S, Kim M, Lee SI, Sohn DH, Robinson WH. Protein phosphatase magnesium-dependent 1A induces inflammation in rheumatoid arthritis. Biochem Biophys Res Commun 2019; 522:731-735. [PMID: 31791585 DOI: 10.1016/j.bbrc.2019.11.112] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 11/17/2019] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is a highly inflammatory autoimmune disease. Although proinflammatory cytokines, including tumor necrosis factor (TNF) and interleukin (IL)-6, play a key role in the pathogenesis of RA, the causes of chronic inflammation are not fully understood. Here, we report that protein phosphatase magnesium-dependent 1A (PPM1A) levels were increased in RA synovial fluid compared with osteoarthritis (OA) synovial fluid and positively correlated with TNF levels. In addition, PPM1A expression was increased in synovial tissue from RA patients and joint tissue from a mouse model of arthritis. Finally, extracellular PPM1A induced inflammation by stimulating macrophages to produce TNF through toll-like receptor 4 (TLR4) and myeloid differentiation primary response protein 88 (MyD88) signaling pathway. Our findings suggest that extracellular PPM1A may contribute to the pathogenesis of RA by functioning as a damage-associated molecular pattern (DAMP) to induce inflammation.
Collapse
Affiliation(s)
- Beomgu Lee
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - You Seon Song
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea; Department of Radiology, Pusan National University Hospital, Busan, Republic of Korea
| | - Christopher Rhodes
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Tae Sik Goh
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea; Department of Orthopaedic Surgery, Pusan National University Hospital, Busan, Republic of Korea
| | - Jong Seong Roh
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Hoim Jeong
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jisu Park
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Han-Na Lee
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Pusan National University School of Medicine, Pusan National University Hospital, Busan, Republic of Korea
| | - Seung-Geun Lee
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Pusan National University School of Medicine, Pusan National University Hospital, Busan, Republic of Korea
| | - Soohyun Kim
- College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Mingyo Kim
- Division of Rheumatology, Department of Internal Medicine, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
| | - Sang-Il Lee
- Division of Rheumatology, Department of Internal Medicine, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea.
| | - William H Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA.
| |
Collapse
|
233
|
Bigaeva E, Gore E, Simon E, Zwick M, Oldenburger A, de Jong KP, Hofker HS, Schlepütz M, Nicklin P, Boersema M, Rippmann JF, Olinga P. Transcriptomic characterization of culture-associated changes in murine and human precision-cut tissue slices. Arch Toxicol 2019; 93:3549-3583. [PMID: 31754732 DOI: 10.1007/s00204-019-02611-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022]
Abstract
Our knowledge of complex pathological mechanisms underlying organ fibrosis is predominantly derived from animal studies. However, relevance of animal models for human disease is limited; therefore, an ex vivo model of human precision-cut tissue slices (PCTS) might become an indispensable tool in fibrosis research and drug development by bridging the animal-human translational gap. This study, presented as two parts, provides comprehensive characterization of the dynamic transcriptional changes in PCTS during culture by RNA sequencing. Part I investigates the differences in culture-induced responses in murine and human PCTS derived from healthy liver, kidney and gut. Part II delineates the molecular processes in cultured human PCTS generated from diseased liver, kidney and ileum. We demonstrated that culture was associated with extensive transcriptional changes and impacted PCTS in a universal way across the organs and two species by triggering an inflammatory response and fibrosis-related extracellular matrix (ECM) remodelling. All PCTS shared mRNA upregulation of IL-11 and ECM-degrading enzymes MMP3 and MMP10. Slice preparation and culturing activated numerous pathways across all PCTS, especially those involved in inflammation (IL-6, IL-8 and HMGB1 signalling) and tissue remodelling (osteoarthritis pathway and integrin signalling). Despite the converging effects of culture, PCTS display species-, organ- and pathology-specific differences in the regulation of genes and canonical pathways. The underlying pathology in human diseased PCTS endures and influences biological processes like cytokine release. Our study reinforces the use of PCTS as an ex vivo fibrosis model and supports future studies towards its validation as a preclinical tool for drug development.
Collapse
Affiliation(s)
- Emilia Bigaeva
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands
| | - Emilia Gore
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands
| | - Eric Simon
- Computational Biology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach an der Riss, Germany
| | - Matthias Zwick
- Computational Biology, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach an der Riss, Germany
| | - Anouk Oldenburger
- Cardiometabolic Disease Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach an der Riss, Germany
| | - Koert P de Jong
- Department of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Hendrik S Hofker
- Department of Surgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Marco Schlepütz
- Respiratory Diseases, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach an der Riss, Germany
| | - Paul Nicklin
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach an der Riss, Germany
| | - Miriam Boersema
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands
| | - Jörg F Rippmann
- Cardiometabolic Disease Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88397, Biberach an der Riss, Germany
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, Groningen, 9713AV, The Netherlands.
| |
Collapse
|
234
|
Budhwar S, Verma P, Verma R, Gupta S, Rai S, Rajender S, Singh K. Altered cord serum 25-hydroxyvitamin D signaling and placental inflammation is associated with pre-term birth. Am J Reprod Immunol 2019; 83:e13201. [PMID: 31642155 DOI: 10.1111/aji.13201] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/30/2019] [Accepted: 10/18/2019] [Indexed: 12/18/2022] Open
Abstract
PROBLEM Vitamin D is well-known for having anti-inflammatory and immunomodulatory properties. Impaired maternal vitamin D status has been known to increase the risk of adverse pregnancy outcomes like pre-term birth. The present study aims to evaluate the impact of fetal cord serum 25-hydroxyvitamin D-mediated signaling in mediating inflammatory responses in placenta during pre-term birth. METHOD OF STUDY For the above purpose, cord serum 25 hydroxyvitamin D 25(OH)D were measured in term (n = 20) and pre-term (n = 20) born babies using ELISA. Vitamin D downstream signaling has also been checked in placenta (VDR, CYP27B1, cathelicidin LL37) along with expression of inflammatory markers (S100A8, HMGB1, TLR2, p-NF-kappaB) using Western blotting and immunohistochemistry. Pearson correlation model was used to do correlation study. RESULTS Compared with term born babies (59.31 ± 3.476), decline in cord serum 25(OH)D levels is observed in pre-term born babies (22.26 ± 1.083, P = <0.0001) that showed strong positive correlation with gestational age (r = .9368***) and birthweight (r = .9559***). On the other hand, vitamin D signaling markers were found to be downregulated and inflammatory markers were upregulated in placental tissue of pre-term born babies. CONCLUSION Thus, our study demonstrated that insufficient cord 25(OH)D levels may disturb the homeostasis of inflammation in placenta. Altered cord serum 25(OH)D mediated anti-inflammatory signaling may be acting as trigger signals in modulating inflammatory responses in placenta and eliciting premature activation of spontaneous labor in pre-term birth.
Collapse
Affiliation(s)
- Snehil Budhwar
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Priyanka Verma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Rachna Verma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Shreshtha Gupta
- Department of Obstetrics and Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sangeeta Rai
- Department of Obstetrics and Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Singh Rajender
- Division of Endocrinology, CSIR Central Drug Research Institute, Lucknow, India
| | - Kiran Singh
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
235
|
Peripheral loss of EphA4 ameliorates TBI-induced neuroinflammation and tissue damage. J Neuroinflammation 2019; 16:210. [PMID: 31711546 PMCID: PMC6844068 DOI: 10.1186/s12974-019-1605-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/26/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The continuum of pro- and anti-inflammatory response elicited by traumatic brain injury (TBI) is suggested to play a key role in the outcome of TBI; however, the underlying mechanisms remain ill -defined. METHODS Here, we demonstrate that using bone marrow chimeric mice and systemic inhibition of EphA4 receptor shifts the pro-inflammatory milieu to pro-resolving following acute TBI. RESULTS EphA4 expression is increased in the injured cortex as early as 2 h post-TBI and on CX3CR1gfp-positive cells in the peri-lesion. Systemic inhibition or genetic deletion of EphA4 significantly reduced cortical lesion volume and shifted the inflammatory profile of peripheral-derived immune cells to pro-resolving in the damaged cortex. These findings were consistent with in vitro studies showing EphA4 inhibition or deletion altered the inflammatory state of LPS-stimulated monocyte/macrophages towards anti-inflammatory. Phosphoarray analysis revealed that EphA4 may regulate pro-inflammatory gene expression by suppressing the mTOR, Akt, and NF-κB pathways. Our human metadata analysis further demonstrates increased EPHA4 and pro-inflammatory gene expression, which correlates with reduced AKT concurrent with increased brain injury severity in patients. CONCLUSIONS Overall, these findings implicate EphA4 as a novel mediator of cortical tissue damage and neuroinflammation following TBI.
Collapse
|
236
|
Chin PY, Dorian C, Sharkey DJ, Hutchinson MR, Rice KC, Moldenhauer LM, Robertson SA. Toll-Like Receptor-4 Antagonist (+)-Naloxone Confers Sexually Dimorphic Protection From Inflammation-Induced Fetal Programming in Mice. Endocrinology 2019; 160:2646-2662. [PMID: 31504393 PMCID: PMC6936318 DOI: 10.1210/en.2019-00493] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 08/21/2019] [Indexed: 12/21/2022]
Abstract
Inflammation elicited by infection or noninfectious insults during gestation induces proinflammatory cytokines that can shift the trajectory of development to alter offspring phenotype, promote adiposity, and increase susceptibility to metabolic disease in later life. In this study, we use mice to investigate the utility of a small molecule Toll-like receptor (TLR)4 antagonist (+)-naloxone, the nonopioid isomer of the opioid receptor antagonist (-)-naloxone, for mitigating altered fetal metabolic programming induced by a modest systemic inflammatory challenge in late gestation. In adult progeny exposed to lipopolysaccharide (LPS) challenge in utero, male but not female offspring exhibited elevated adipose tissue, reduced muscle mass, and elevated plasma leptin at 20 weeks of age. Effects were largely reversed by coadministration of (+)-naloxone following LPS. When given alone without LPS, (+)-naloxone elicited accelerated postweaning growth and elevated muscle and fat mass in adult male but not female offspring. LPS induced expression of inflammatory cytokines Il1a, Il1b, Il6, Tnf, and Il10 in fetal brain, placental, and uterine tissues, and (+)-naloxone suppressed LPS-induced cytokine expression. Fetal sex-specific regulation of cytokine expression was evident, with higher Il1a, Il1b, Il6, and Il10 induced by LPS in tissues associated with male fetuses, and greater suppression by (+)-naloxone of Il6 in females. These data demonstrate that modulating TLR4 signaling with (+)-naloxone provides protection from inflammatory diversion of fetal developmental programming in utero, associated with attenuation of gestational tissue cytokine expression in a fetal sex-specific manner. The results suggest that pharmacologic interventions targeting TLR4 warrant evaluation for attenuating developmental programming effects of fetal exposure to maternal inflammatory mediators.
Collapse
Affiliation(s)
- Peck Yin Chin
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Camilla Dorian
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - David J Sharkey
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Mark R Hutchinson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Adelaide, South Australia, Australia
| | - Kenner C Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland
| | - Lachlan M Moldenhauer
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Correspondence: Sarah A. Robertson, PhD, Robinson Research Institute and the Adelaide Medical School, University of Adelaide, North Terrace, Adelaide, South Australia 5005, Australia. E-mail:
| |
Collapse
|
237
|
de Leve S, Wirsdörfer F, Jendrossek V. The CD73/Ado System-A New Player in RT Induced Adverse Late Effects. Cancers (Basel) 2019; 11:cancers11101578. [PMID: 31623231 PMCID: PMC6827091 DOI: 10.3390/cancers11101578] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/11/2019] [Accepted: 10/12/2019] [Indexed: 02/06/2023] Open
Abstract
Radiotherapy (RT) is a central component of standard treatment for many cancer patients. RT alone or in multimodal treatment strategies has a documented contribution to enhanced local control and overall survival of cancer patients, and cancer cure. Clinical RT aims at maximizing tumor control, while minimizing the risk for RT-induced adverse late effects. However, acute and late toxicities of IR in normal tissues are still important biological barriers to successful RT: While curative RT may not be tolerable, sub-optimal tolerable RT doses will lead to fatal outcomes by local recurrence or metastatic disease, even when accepting adverse normal tissue effects that decrease the quality of life of irradiated cancer patients. Technical improvements in treatment planning and the increasing use of particle therapy have allowed for a more accurate delivery of IR to the tumor volume and have thereby helped to improve the safety profile of RT for many solid tumors. With these technical and physical strategies reaching their natural limits, current research for improving the therapeutic gain of RT focuses on innovative biological concepts that either selectively limit the adverse effects of RT in normal tissues without protecting the tumor or specifically increase the radiosensitivity of the tumor tissue without enhancing the risk of normal tissue complications. The biology-based optimization of RT requires the identification of biological factors that are linked to differential radiosensitivity of normal or tumor tissues, and are amenable to therapeutic targeting. Extracellular adenosine is an endogenous mediator critical to the maintenance of homeostasis in various tissues. Adenosine is either released from stressed or injured cells or generated from extracellular adenine nucleotides by the concerted action of the ectoenzymes ectoapyrase (CD39) and 5′ ectonucleotidase (NT5E, CD73) that catabolize ATP to adenosine. Recent work revealed a role of the immunoregulatory CD73/adenosine system in radiation-induced fibrotic disease in normal tissues suggesting a potential use as novel therapeutic target for normal tissue protection. The present review summarizes relevant findings on the pathologic roles of CD73 and adenosine in radiation-induced fibrosis in different organs (lung, skin, gut, and kidney) that have been obtained in preclinical models and proposes a refined model of radiation-induced normal tissue toxicity including the disease-promoting effects of radiation-induced activation of CD73/adenosine signaling in the irradiated tissue environment. However, expression and activity of the CD73/adenosine system in the tumor environment has also been linked to increased tumor growth and tumor immune escape, at least in preclinical models. Therefore, we will discuss the use of pharmacologic inhibition of CD73/adenosine-signaling as a promising strategy for improving the therapeutic gain of RT by targeting both, malignant tumor growth and adverse late effects of RT with a focus on fibrotic disease. The consideration of the therapeutic window is particularly important in view of the increasing use of RT in combination with various molecularly targeted agents and immunotherapy to enhance the tumor radiation response, as such combinations may result in increased or novel toxicities, as well as the increasing number of cancer survivors.
Collapse
Affiliation(s)
- Simone de Leve
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| | - Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| |
Collapse
|
238
|
Yang J, Zhao Y, Zhang L, Fan H, Qi C, Zhang K, Liu X, Fei L, Chen S, Wang M, Kuang F, Wang Y, Wu S. RIPK3/MLKL-Mediated Neuronal Necroptosis Modulates the M1/M2 Polarization of Microglia/Macrophages in the Ischemic Cortex. Cereb Cortex 2019; 28:2622-2635. [PMID: 29746630 PMCID: PMC5998990 DOI: 10.1093/cercor/bhy089] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Indexed: 12/28/2022] Open
Abstract
Cell death and subsequent inflammation are 2 key pathological changes occurring in cerebral ischemia. Active microglia/macrophages play a double-edged role depending on the balance of their M1/M2 phenotypes. Necrosis is the predominant type of cell death following ischemia. However, how necrotic cells modulate the M1/M2 polarization of microglia/macrophages remains poorly investigated. Here, we reported that ischemia induces a rapid RIPK3/MLKL-mediated neuron-dominated necroptosis, a type of programmed necrosis. Ablating RIPK3 or MLKL could switch the activation of microglia/macrophages from M1 to the M2 type in the ischemic cortex. Conditioned medium of oxygen-glucose deprivation (OGD)-treated wild-type (WT) neurons induced M1 polarization, while that of RIPK3−/− neurons favored M2 polarization. OGD treatment induces proinflammatory IL-18 and TNFα in WT but not in RIPK3−/− neurons, which in turn upregulate anti-inflammatory IL-4 and IL-10. Furthermore, the expression of Myd88—a common downstream adaptor of toll-like receptors—is significantly upregulated in the microglia/macrophages of ischemic WT but not of RIPK3−/− or MLKL−/− cortices. Antagonizing the function of Myd88 could phenocopy the effects of RIPK3/MLKL-knockout on the polarization of microglia/macrophages and was neuroprotective. Our data revealed a novel role of necroptotic neurons in modulating the M1/M2 balance of microglia/macrophages in the ischemic cortex, possibly through Myd88 signaling.
Collapse
Affiliation(s)
- Jiping Yang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China.,Department of Anatomy, Shaanxi Key Laboratory of Brain Disorders and Institute of Basic Medical Sciences, Xi'an Medical University, 1 Xin Wang Road, Xi'an, Shaanxi, China
| | - Youyi Zhao
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China.,School of Basic Medicine, Chengdu Medical College, Chengdu, China
| | - Li Zhang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China.,Department of Anatomy, Shaanxi Key Laboratory of Brain Disorders and Institute of Basic Medical Sciences, Xi'an Medical University, 1 Xin Wang Road, Xi'an, Shaanxi, China
| | - Hong Fan
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China
| | - Chuchu Qi
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China
| | - Kun Zhang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China
| | - Xinyu Liu
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China
| | - Lin Fei
- Department of Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yan Ta Western Road, Xi'an, Shaanxi, China
| | - Siwei Chen
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China
| | - Mengmeng Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China
| | - Fang Kuang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China
| | - Yazhou Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China
| | - Shengxi Wu
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, Xi'an, Shaanxi, China
| |
Collapse
|
239
|
Hong Y, Kim YK, Kim GB, Nam GH, Kim SA, Park Y, Yang Y, Kim IS. Degradation of tumour stromal hyaluronan by small extracellular vesicle-PH20 stimulates CD103 + dendritic cells and in combination with PD-L1 blockade boosts anti-tumour immunity. J Extracell Vesicles 2019; 8:1670893. [PMID: 31632619 PMCID: PMC6781230 DOI: 10.1080/20013078.2019.1670893] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/21/2019] [Accepted: 09/14/2019] [Indexed: 12/13/2022] Open
Abstract
Highly accumulated hyaluronan (HA) not only provides a physiological barrier but also supports an immune-suppressive tumour microenvironment. High-molecular-weight (HMW)-HA inhibits the activation of immune cells and their access into tumour tissues, whereas, low-molecular-weight oligo-HA is known to potentially activate dendritic cells (DCs). In this paper, we investigated whether small extracellular vesicle (EVs)-PH20 hyaluronidase induces tumour HA degradation, which, in turn, activates DCs to promote anti-cancer immune responses. Informed by our previous work, we used a small EV carrying GPI-anchored PH20 hyaluronidase (Exo-PH20) that could deeply penetrate into tumour foci via HA degradation. We found that Exo-PH20-treatment successfully activates the maturation and migration of DCs in vivo, particularly CD103+ DCs leading to the activation of tumour-specific CD8+ T cells, which work together to inhibit tumour growth. Moreover, combination with anti-PD-L1 antibody provided potent tumour-specific CD8+ T cell immune responses as well as elicited prominent tumour growth inhibition both in syngenic and spontaneous breast cancer models, and this anti-tumour immunity was durable. Together, these results present new insights for HA degradation by Exo-PH20, providing a better understanding of oligo HA-triggered immune responses to cancer.
Collapse
Affiliation(s)
- Yeonsun Hong
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Yoon Kyoung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Gi Beom Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Gi-Hoon Nam
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Yoon Park
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Yoosoo Yang
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| |
Collapse
|
240
|
Paudel YN, Angelopoulou E, Piperi C, Balasubramaniam VR, Othman I, Shaikh MF. Enlightening the role of high mobility group box 1 (HMGB1) in inflammation: Updates on receptor signalling. Eur J Pharmacol 2019; 858:172487. [DOI: 10.1016/j.ejphar.2019.172487] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/19/2019] [Accepted: 06/19/2019] [Indexed: 12/17/2022]
|
241
|
Buckenmeyer MJ, Meder TJ, Prest TA, Brown BN. Decellularization techniques and their applications for the repair and regeneration of the nervous system. Methods 2019; 171:41-61. [PMID: 31398392 DOI: 10.1016/j.ymeth.2019.07.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/11/2019] [Accepted: 07/26/2019] [Indexed: 01/15/2023] Open
Abstract
A variety of surgical and non-surgical approaches have been used to address the impacts of nervous system injuries, which can lead to either impairment or a complete loss of function for affected patients. The inherent ability of nervous tissues to repair and/or regenerate is dampened due to irreversible changes that occur within the tissue remodeling microenvironment following injury. Specifically, dysregulation of the extracellular matrix (i.e., scarring) has been suggested as one of the major factors that can directly impair normal cell function and could significantly alter the regenerative potential of these tissues. A number of tissue engineering and regenerative medicine-based approaches have been suggested to intervene in the process of remodeling which occurs following injury. Decellularization has become an increasingly popular technique used to obtain acellular scaffolds, and their derivatives (hydrogels, etc.), which retain tissue-specific components, including critical structural and functional proteins. These advantageous characteristics make this approach an intriguing option for creating materials capable of stimulating the sensitive repair mechanisms associated with nervous system injuries. Over the past decade, several diverse decellularization methods have been implemented specifically for nervous system applications in an attempt to carefully remove cellular content while preserving tissue morphology and composition. Each application-based decellularized ECM product requires carefully designed treatments that preserve the unique biochemical signatures associated within each tissue type to stimulate the repair of brain, spinal cord, and peripheral nerve tissues. Herein, we review the decellularization techniques that have been applied to create biomaterials with the potential to promote the repair and regeneration of tissues within the central and peripheral nervous system.
Collapse
Affiliation(s)
- Michael J Buckenmeyer
- Department of Bioengineering, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States.
| | - Tyler J Meder
- Department of Bioengineering, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States.
| | - Travis A Prest
- Department of Bioengineering, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States.
| | - Bryan N Brown
- Department of Bioengineering, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States; Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, United States.
| |
Collapse
|
242
|
Chen C, Gao R, Li M, Wang Q, Chen H, Zhang S, Mao X, Behensky A, Zhang Z, Gan L, Li T, Liao R, Li Q, Yu H, Yang J, Zhu T, Liu J. Extracellular RNAs-TLR3 signaling contributes to cognitive decline in a mouse model of postoperative cognitive dysfunction. Brain Behav Immun 2019; 80:439-451. [PMID: 30980952 DOI: 10.1016/j.bbi.2019.04.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 03/23/2019] [Accepted: 04/09/2019] [Indexed: 01/28/2023] Open
Abstract
Postoperative cognitive dysfunction (POCD) is considered a severe complication after surgery among elderly patients. Toll-like receptor 3 (TLR3) has recently been reported to play an important role in hippocampus-dependent working memory. However, the role of TLR3 in the development of POCD remains unclear. In the current study, we hypothesized that increased extracellular RNAs (exRNAs) during anesthesia and surgical operation, especially double stranded RNAs (dsRNAs), would activate TLR3 signaling pathways and mediate POCD. Using a mouse model of POCD, 20-22 months wild-type (WT) mice were undergoing unilateral nephrectomy and increased TLR3 expression levels and co-localization with neuronal and microglial cells were found in the surgery group compared with the sham group. Compared with WT mice, TLR3 knockout (KO, -/-) mice had improved hippocampus-dependent memory and attenuated production of inflammatory cytokines and apoptosis. Increased exRNAs and/or co-localization with TLR3 were found in both in vitro and in vivo models. Of note, TLR3/dsRNA complex inhibitor administration reduced hippocampal dsRNA level and TLR3 expression, attenuated hippocampal inflammatory cytokines production and apoptosis, and thus improved hippocampus-dependent memory. Our results indicate that exRNAs, especially dsRNAs, present under stressful conditions may trigger TLR3 activation and initiate the downstream inflammatory and apoptotic signaling, and play a substantial role in the development of POCD.
Collapse
Affiliation(s)
- Chan Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Rui Gao
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ming Li
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qiao Wang
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hai Chen
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shu Zhang
- Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiaobo Mao
- Institute of Cell Engineering, Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Adam Behensky
- Institute of Cell Engineering, Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Zheng Zhang
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lu Gan
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Tao Li
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ren Liao
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qian Li
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hai Yu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jing Yang
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Tao Zhu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Jin Liu
- Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
243
|
Peng A, Lu X, Huang J, He M, Xu J, Huang H, Chen Q. Rheumatoid arthritis synovial fibroblasts promote TREM-1 expression in monocytes via COX-2/PGE 2 pathway. Arthritis Res Ther 2019; 21:169. [PMID: 31287012 PMCID: PMC6615166 DOI: 10.1186/s13075-019-1954-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 06/28/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Triggering receptor expressed on myeloid cells-1 (TREM-1) is inducible on monocyte/macrophages and neutrophils and amplifies the inflammatory response. The aim of this study was to determine whether rheumatoid arthritis synovial fibroblasts (RASF) promote the expression of TREM-1 in monocytes and its potential regulatory mechanism. METHODS Synovial fluid and paired peripheral blood from rheumatoid arthritis (RA) patients were analyzed using flow cytometry. Expression of TREM-1 in monocytes was detected after co-culture with RASF, with or without pre-treatment with toll-like receptor (TLR) ligands. Whether RASF-regulated TREM-1 level in monocytes require direct cell contact or soluble factors was evaluated by transwell experiment. COX-2 expression and PGE2 secretion in RASF were determined by quantitative PCR (qPCR) and ELISA. RASF, with and without TLR ligand stimulation, were treated with COX-2 inhibitors, COX-2 siRNA (siCOX-2) or EP1-4 antagonists, and the resulting TREM-1 level in CD14+ monocytes was measured using flow cytometry. RESULTS TREM-1 was highly expressed in CD14+ cells from peripheral blood and especially synovial fluid from RA patients. The expression of TREM-1 in monocytes was increased by co-culture with RASF. TLR-ligand-activated RASF further elevated TREM-1 level. Transwell assay indicated that soluble factors played a key role in RASF-promoted expression of TREM-1 in monocytes. RASF, with or without stimulation by TLR ligands, increased secretion of PGE2 in a cyclooxygenase (COX)-2-dependent manner. PGE2 enhanced the increase in TREM-1 level in monocytes. Finally, studies using COX-2 inhibitors, COX-2 siRNA (siCOX-2) and EP1-4 antagonists, showed that RASF promotion of TREM-1 expression in monocytes was mediated by COX-2/PGE2/EP2,4 signaling. CONCLUSIONS Our data is the first report to reveal the critical role of RASF in upregulating TREM-1 expression in monocytes, which indicates that TREM-1 might be a novel target for RA therapy.
Collapse
Affiliation(s)
- Anping Peng
- Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Xinyi Lu
- Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jun Huang
- Department of Pathogenic Biology and Immunology, Guangzhou Medical University, Guangzhou, China
| | - Min He
- Department of Laboratory Science, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhua Xu
- Department of Laboratory Science, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui Huang
- Department of Laboratory Science, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qubo Chen
- Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
244
|
Brajer-Luftmann B, Nowicka A, Kaczmarek M, Wyrzykiewicz M, Yasar S, Piorunek T, Sikora J, Batura-Gabryel H. Damage-Associated Molecular Patterns and Myeloid-Derived Suppressor Cells in Bronchoalveolar Lavage Fluid in Chronic Obstructive Pulmonary Disease Patients. J Immunol Res 2019; 2019:9708769. [PMID: 31355298 PMCID: PMC6636560 DOI: 10.1155/2019/9708769] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 04/11/2019] [Accepted: 05/14/2019] [Indexed: 02/07/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are present in the human lung microenvironment, and they may be involved in the local inflammatory process in chronic obstructive pulmonary disease (COPD). Chronic inflammation in COPD may induce immunogenic cell death of structural airway cells, causing the release of damage-associated molecular patterns (DAMPs). DAMPs may activate the innate and adaptive immune system. The relationship between MDSCs and DAMPs in COPD is poorly described in the available literature. Objectives. (1) Assessment of MDSC percentage and DAMP concentration in bronchoalveolar lavage fluid (BALF) and peripheral blood. (2) Analysis of the relationship between MDSC percentage and chosen DAMPs. Patients and Methods. 30 COPD patients were included. Using monoclonal antibodies directly conjugated with fluorochromes in flow cytometry, MDSCs were assessed in BALF and peripheral blood. The concentration of DAMPs was estimated using sandwich ELISA. Using the Bradford method, the total protein concentrations were evaluated. Results. The percentage of MDSCs among MC in BALF correlated well with the concentration of defensin and heat shock protein 27. Assessing the percentage of MDSCs among all leukocytes in BALF, we revealed a significant correlation with the concentration of defensin, hyaluronic acid, and surfactant protein A. No dependencies occurred between DAMPs and MDSCs in peripheral blood. Conclusion. MDSCs and DAMPs occur in the COPD patient lung microenvironment. Significant correlations between them found in BALF may indicate their influence on the local inflammatory process in COPD. These relationships allow better understanding of the inflammatory process in COPD.
Collapse
Affiliation(s)
- Beata Brajer-Luftmann
- Department of Pulmonology, Allergology and Pulmonary Oncology, Poznan University of Medical Sciences, Szamarzewskiego 84 Street, 60-569 Poznan, Poland
| | - Agata Nowicka
- Department of Pulmonology, Allergology and Pulmonary Oncology, Poznan University of Medical Sciences, Szamarzewskiego 84 Street, 60-569 Poznan, Poland
| | - Mariusz Kaczmarek
- Department of Clinical Immunology, Poznan University of Medical Sciences, Rokietnicka 5D Street, 60-806 Poznan, Poland
| | - Magdalena Wyrzykiewicz
- Department of Clinical Immunology, Poznan University of Medical Sciences, Rokietnicka 5D Street, 60-806 Poznan, Poland
| | - Senan Yasar
- Department of Clinical Immunology, Poznan University of Medical Sciences, Rokietnicka 5D Street, 60-806 Poznan, Poland
| | - Tomasz Piorunek
- Department of Pulmonology, Allergology and Pulmonary Oncology, Poznan University of Medical Sciences, Szamarzewskiego 84 Street, 60-569 Poznan, Poland
| | - Jan Sikora
- Department of Clinical Immunology, Poznan University of Medical Sciences, Rokietnicka 5D Street, 60-806 Poznan, Poland
| | - Halina Batura-Gabryel
- Department of Pulmonology, Allergology and Pulmonary Oncology, Poznan University of Medical Sciences, Szamarzewskiego 84 Street, 60-569 Poznan, Poland
| |
Collapse
|
245
|
Choroid plexus transcriptome and ultrastructure analysis reveals a TLR2-specific chemotaxis signature and cytoskeleton remodeling in leukocyte trafficking. Brain Behav Immun 2019; 79:216-227. [PMID: 30822467 PMCID: PMC6591031 DOI: 10.1016/j.bbi.2019.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 01/09/2019] [Accepted: 02/06/2019] [Indexed: 01/15/2023] Open
Abstract
Perinatal infection and inflammation are major risk factors for injury in the developing brain, however, underlying mechanisms are not fully understood. Leukocyte migration to the cerebrospinal fluid (CSF) and brain is a hallmark of many pathologies of the central nervous system including those in neonates. We previously reported that systemic activation of Toll-like receptor (TLR) 2, a major receptor for gram-positive bacteria, by agonist Pam3CSK4 (P3C) resulted in dramatic neutrophil and monocyte infiltration to the CSF and periventricular brain of neonatal mice, an effect that was absent by the TLR4 agonist, LPS. Here we first report that choroid plexus is a route of TLR2-mediated leukocyte infiltration to the CSF by performing flow cytometry and transmission electron microscopy (TEM) of the choroid plexus. Next, we exploited the striking discrepancy between P3C and LPS effects on cell migration to determine the pathways regulating leukocyte trafficking through the choroid plexus. We performed RNA sequencing on the choroid plexus after administration of P3C and LPS to postnatal day 8 mice. A cluster gene analysis revealed a TLR2-specific signature of chemotaxis represented by 80-fold increased expression of the gene Ccl3 and 1000-fold increased expression of the gene Cxcl2. Ingenuity pathway analysis (IPA) revealed TLR2-specific molecular signaling related to cytoskeleton organization (e.g. actin signaling) as well as inositol phospholipids biosynthesis and degradation. This included upregulation of genes such as Rac2 and Micall2. In support of IPA results, ultrastructural analysis by TEM revealed clefting and perforations in the basement membrane of the choroid plexus epithelial cells in P3C-treated mice. In summary, we show that the choroid plexus is a route of TLR2-mediated transmigration of neutrophils and monocytes to the developing brain, and reveal previously unrecognized mechanisms that includes a specific chemotaxis profile as well as pathways regulating cytoskeleton and basement membrane remodeling.
Collapse
|
246
|
Chen Z, Liu Q, Zhu Z, Xiang F, Wu R, Kang X. Toll-like receptor 4 contributes to uterine activation by upregulating pro-inflammatory cytokine and CAP expression via the NF-κB/P38MAPK signaling pathway during pregnancy. J Cell Physiol 2019; 235:513-525. [PMID: 31236964 DOI: 10.1002/jcp.28991] [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: 04/08/2019] [Accepted: 05/29/2019] [Indexed: 11/06/2022]
Abstract
Evidence indicates that inflammatory response is significant during the physiological process of human parturition; however, the specific signaling pathway that triggers inflammation is undefined. Toll-like receptors (TLRs) are key upstream gatekeepers that control inflammatory activation before preterm delivery. Our previous study showed that TLR4 expression was significantly increased in human pregnancy tissue during preterm and term labor. Therefore, we explore whether TLR4 plays a role in term labor by initiating inflammatory responses, therefore promoting uterine activation. The results showed that expression of TLR4, interleukin-1β (IL-1β), IL-6, tumor necrosis factor-α (TNF-α), CC chemokine ligand 2 (CCL-2), and uterine contraction-associated proteins (CAPs) was upregulated in the human and mice term labor (TL) group compared with the not-in-labor (TNL) group, and the TLR4 level positively correlated with CAP expression. In pregnant TLR4-knockout (TLR4-/- ) mice, gestation length was extended by 8 hr compared with the wild-type group, and the expression of IL-1β, IL-6, TNF-α, CCL-2, and CAPs was decreased in TLR4-/- mice. Furthermore, nuclear factor-κB (NF-κB) and P38MAPK activation is involved in the initiation of labor but was inhibited in TLR4-/- mice. In uterine smooth muscle cells, the expression of inflammatory cytokines and CAPs decreased when the NF-κB and P38MAPK pathway was inhibited. Our data suggest that TLR4 is a key factor in regulating the inflammatory response that drives uterine activation and delivery initiation via activating the NF-κB/P38MAPK pathway.
Collapse
Affiliation(s)
- Zixi Chen
- Department of Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiaoli Liu
- Department of Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhaowei Zhu
- Department of Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fenfen Xiang
- Department of Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Wu
- Department of Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiangdong Kang
- Department of Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
247
|
Nascimento LFMD, Moura LDD, Lima RT, Cruz MDSPE. Novos adjuvantes vacinais: importante ferramenta para imunoterapia da leishmaniose visceral. HU REVISTA 2019. [DOI: 10.34019/1982-8047.2018.v44.14123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Atualmente, muitas das vacinas em desenvolvimento são aquelas compostas de proteínas antigênicas individuais de parasitas ou uma combinação de vários antígenos individuais que são produzidos como produtos recombinantes obtidos por técnicas de biologia molecular. Dentre elas a Leish-111f e sua variação Leish-110f tem ganhado destaque na proteção contra a LV e LC e alcançaram estudos de fase II em seres humanos. A eficácia de uma vacina é otimizada pela adição de adjuvantes imunológicos. No entanto, embora os adjuvantes tenham sido usados por mais de um século, até o momento, apenas alguns adjuvantes são aprovados para o uso em humanos, a maioria destinada a melhorar a eficácia da vacina e a produção de anticorpos protetores específicos do antígeno. Os mecanismos de ação dos adjuvantes imunológicos são diversos, dependendo da sua natureza química e molecular sendo capazes de ativar células imunes especificas que conduzem a respostas imunes inatas e adaptativas melhoradas. Embora o mecanismo de ação molecular detalhado de muitos adjuvantes ainda seja desconhecido, a descoberta de receptores Toll-like (TLRs) forneceu informações críticas sobre o efeito imunoestimulador de numerosos componentes bacterianos que envolvem interação com receptores TLRs, mostrando que estes ligantes melhoram tanto a qualidade como a quantidade de respostas imunes adaptativas do hospedeiro quando utilizadas em formulações de vacinais direcionadas para doenças. O potencial desses adjuvantes de TLR em melhorar o design e os resultados de várias vacinas está em constante evolução, à medida que novos agonistas são descobertos e testados em modelos experimentais e estudos clínicos de vacinação. Nesta revisão, é apresentado um resumo do progresso recente no desenvolvimento de proteínas recombinantes de segunda geração e adjuvantes de TLR, sendo o foco principal nos TLR4 e suas melhorias.
Collapse
|
248
|
Vancsik T, Forika G, Balogh A, Kiss E, Krenacs T. Modulated electro-hyperthermia induced p53 driven apoptosis and cell cycle arrest additively support doxorubicin chemotherapy of colorectal cancer in vitro. Cancer Med 2019; 8:4292-4303. [PMID: 31183995 PMCID: PMC6675742 DOI: 10.1002/cam4.2330] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 03/20/2019] [Accepted: 03/20/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Modulated electro-hyperthermia (mEHT), a noninvasive complementary treatment of human chemo- and radiotherapy, can generate selective ~42°C heat in cancer due to elevated glycolysis (Warburg-effect) and electric conductivity in malignant tissues. Here we tested the molecular background of mEHT and its combination with doxorubicin chemotherapy using an in vitro model. METHODS C26 mouse colorectal adenocarcinoma cultures were mEHT treated at 42°C for 2 × 60 minutes (with 120 minutes interruption) either alone or in combination with 1 µmol/L doxorubicin (mEHT + Dox). Cell stress response, apoptosis, and cell cycle regulation related markers were detected using qPCR and immunocytochemistry supported with resazurin cell viability assay, cell death analysis using flow-cytometry and clonogenic assay. RESULT Cell-stress by mEHT alone was indicated by the significant upregulation and release of hsp70 and calreticulin proteins 3 hours posttreatment. Between 3 and 9 hours after treatment significantly reduced anti-apoptotic XIAP, BCL-2, and BCL-XL and elevated pro-apoptotic BAX and PUMA, as well as the cyclin dependent kinase inhibitor p21waf1 mRNA levels were detected. After 24 hours, major elevation and nuclear translocation of phospho-p53(Ser15) protein levels and reduced phospho-Akt(Ser473) levels were accompanied by a significant caspase-3-mediated programmed cell death response. While mEHT dominantly induced apoptosis, Dox administration primarily led to tumor cell necrosis, and both significantly reduced the number of tumor progenitor colonies 10 days post-treatment. Furthermore, mEHT promoted the uptake of Dox by tumor cells and the combined treatment additively reduced tumor cell viability and augmented cell death near to synergy. CONCLUSION In C26 colorectal adenocarcinoma mEHT-induced irreversible cell stress can activate both caspase-dependent apoptosis and p21waf1 mediated growth arrest pathways, likely to be driven by the upregulated nuclear p53 protein. Elevated phospho-p53(Ser15) might contribute to p53 escape from mdm2 control, which was further supported by reduced phospho-Akt(Ser473) protein levels. In combinations, mEHT could promote the uptake and significantly potentiate the cytotoxic effect of doxorubicin.
Collapse
Affiliation(s)
- Tamas Vancsik
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gertrud Forika
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Andrea Balogh
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Eva Kiss
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Tibor Krenacs
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
249
|
Martinez-Quinones P, Komic A, McCarthy CG, Webb RC, Wenceslau CF. Targeting Endothelial Barrier Dysfunction Caused by Circulating Bacterial and Mitochondrial N-Formyl Peptides With Deformylase. Front Immunol 2019; 10:1270. [PMID: 31244835 PMCID: PMC6563851 DOI: 10.3389/fimmu.2019.01270] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 05/17/2019] [Indexed: 01/05/2023] Open
Abstract
Despite recent advances in our understanding of the mechanisms underlying systemic inflammatory response syndrome (SIRS) and sepsis, the current therapeutic approach to these critically ill patients is centered around supportive care including fluid resuscitation, vasopressors and source control. The incidence of SIRS and sepsis continues to increase in the United States and patients die due to failure to respond to the traditional therapies of nitric oxide blockade, adrenergic agonists, etc. Bacterial and mitochondrial N-formyl peptides (NFPs) act as damage-associated molecular patterns and activate the innate immune system through formyl peptide receptors (FPR) located in immune and non-immune cells, including the vascular endothelium. The resulting inflammatory response manifests as capillary leak, tissue hypoperfusion and vasoplegia, partially due to endothelium barrier breakdown. Potential strategies to prevent this response include decreasing NFP release, breakdown of NFPs, and blocking NFPs from binding FPR. We propose the use of deformylase, the degrading enzyme for NFPs, as potential therapeutic approach to prevent the deleterious effects of NFPs in SIRS and sepsis.
Collapse
Affiliation(s)
- Patricia Martinez-Quinones
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Amel Komic
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, United States
| | - R Clinton Webb
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | | |
Collapse
|
250
|
Igata M, Islam MA, Tada A, Takagi M, Kober AKMH, Albarracin L, Aso H, Ikeda-Ohtsubo W, Miyazawa K, Yoda K, He F, Takahashi H, Villena J, Kitazawa H. Transcriptome Modifications in Porcine Adipocytes via Toll-Like Receptors Activation. Front Immunol 2019; 10:1180. [PMID: 31191544 PMCID: PMC6549529 DOI: 10.3389/fimmu.2019.01180] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/09/2019] [Indexed: 12/15/2022] Open
Abstract
Adipocytes are the most important cell type in adipose tissue playing key roles in immunometabolism. We previously reported that nine members of the Toll-like receptor (TLR) family are expressed in an originally established porcine intramuscular pre-adipocyte (PPI) cell line. However, the ability of TLR ligands to modulate immunometabolic transcriptome modifications in porcine adipocytes has not been elucidated. Herein, we characterized the global transcriptome modifications in porcine intramuscular mature adipocytes (pMA), differentiated from PPI, following stimulation with Pam3csk4, Poly(I:C) or LPS which are ligands for TLR2, TLR3, and TLR4, respectively. Analysis of microarray data identified 530 (218 up, 312 down), 520 (245 up, 275 down), and 525 (239 up, 286 down) differentially expressed genes (DEGs) in pMA following the stimulation with Pam3csk4, Poly(I:C), and LPS, respectively. Gene ontology classification revealed that DEGs are involved in several biological processes including those belonging to immune response and lipid metabolism pathways. Functionally annotated genes were organized into two groups for downstream analysis: immune response related genes (cytokines, chemokines, complement factors, adhesion molecules, and signal transduction), and genes involved with metabolic and endocrine functions (hormones and receptors, growth factors, and lipid biosynthesis). Differential expression analysis revealed that EGR1, NOTCH1, NOS2, TNFAIP3, TRAF3IP1, INSR, CXCR4, PPARA, MAPK10, and C3 are the top 10 commonly altered genes of TLRs induced transcriptional modification of pMA. However, the protein-protein interaction network of DEGs identified EPOR, C3, STAR, CCL2, and SAA2 as the major hub genes, which were also exhibited higher centrality estimates in the Gene-Transcription factor interaction network. Our results provide new insights of transcriptome modifications associated with TLRs activation in porcine adipocytes and identified key regulatory genes that could be used as biomarkers for the evaluation of treatments having immunomodularoty and/or metabolic functional beneficial effects in porcine adipocytes.
Collapse
Affiliation(s)
- Manami Igata
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Md Aminul Islam
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Department of Medicine, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Asuka Tada
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Michihiro Takagi
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - A K M Humayun Kober
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Department of Dairy and Poultry Science, Chittagong Veterinary and Animal Sciences University, Chittangong, Bangladesh
| | - Leonardo Albarracin
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán, Argentina.,Scientific Computing Laboratory, Computer Science Department, Faculty of Exact Science and Technology, National University of Tucuman, San Miguel de Tucumán, Argentina
| | - Hisashi Aso
- Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Cell Biology Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Wakako Ikeda-Ohtsubo
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Kenji Miyazawa
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., Yokohama, Japan
| | - Kazutoyo Yoda
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., Yokohama, Japan
| | - Fang He
- Technical Research Laboratory, Takanashi Milk Products Co., Ltd., Yokohama, Japan
| | - Hideki Takahashi
- Laboratory of Plant Pathology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Plant Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Julio Villena
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), San Miguel de Tucumán, Argentina
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Products Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan.,Livestock Immunology Unit, International Education and Research Centre for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|