1
|
Škorjanc A, Smrkolj V, Umek N. GOReverseLookup: A gene ontology reverse lookup tool. Comput Biol Med 2025; 191:110185. [PMID: 40239235 DOI: 10.1016/j.compbiomed.2025.110185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 03/27/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND AND OBJECTIVE The Gene Ontology (GO) project has been pivotal in providing a structured framework for characterizing genes and annotating them to specific biological concepts. While traditional gene annotation primarily focuses on mapping genes to GO terms, descriptors of biological concepts, there is a growing need for tools facilitating reverse querying. This paper introduces GOReverseLookup, a novel tool designed to identify over- or underrepresented genes in researcher-defined states of interest (phenotypes), described by sets of GO terms. GOReverseLookup supplements the existing power of Gene Ontology by the possibility of orthologous gene querying across several databases, such as Ensembl and UniProtKB. This combination allows for a more nuanced identification of significant genes across a range of cross-species research contexts. METHODS GOReverseLookup queries genes associated with input GO terms. Bundles of GO terms encapsulate user-defined states of interest, e.g., angiogenesis. In the second stage of the analysis, all GO terms associated with each gene are fetched, and finally, the statistical relevance of the genes being involved in one (or all) of the defined states of interests is computed. RESULTS The two presented use cases illustrate its utility in discovering genes related to rheumatoid arthritis and genes linked with chronic inflammation and tumorigenesis. In both cases, GOReverseLookup discovered a substantial number of genes significantly associated with the aforementioned states of interest. CONCLUSIONS GOReverseLookup proves to be a valuable resource for unraveling the genetic basis of phenotypes, with diverse practical potentials in functional genomics, systems biology, and drug discovery. We anticipate that GOReverseLookup will significantly aid in identifying potential gene targets during the initial research phases.
Collapse
Affiliation(s)
- Aljoša Škorjanc
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia
| | - Vladimir Smrkolj
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia; National Institute of Chemistry, Hajdrihova ulica 19, Ljubljana, Slovenia
| | - Nejc Umek
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Korytkova 2, Ljubljana, Slovenia.
| |
Collapse
|
2
|
Cros A, Segura E. IL1R2 Acts as a Negative Regulator of Monocyte Recruitment During Inflammation. Eur J Immunol 2025; 55:e202451468. [PMID: 39610166 PMCID: PMC11739673 DOI: 10.1002/eji.202451468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024]
Abstract
IL1-β plays a central role in inflammation but its biological action needs to be tightly controlled. Such negative regulation can be exerted by the decoy receptor IL1R2. However, IL1R2 biology in immune cells remains poorly characterized, in particular in monocytes. Using conditional deficient mice, we show that Il1r2 deficiency in monocytes does not affect their steady-state life cycle but dysregulates their trafficking to inflamed tissues in models of peritonitis and neuro-inflammation. Mechanistically, we found that Il1r2 deficiency in monocytes increases CCL2 secretion in the inflamed peritoneum, thereby amplifying monocyte recruitment from blood. In autoimmune neuro-inflammation, Il1r2 deficiency in monocytes exacerbates disease severity. Our findings suggest that the specific action of IL1R2 in monocytes contributes to a feedback mechanism for fine-tuning the numbers of recruited monocytes during inflammation.
Collapse
Affiliation(s)
- Adeline Cros
- Institut Curie, PSL Research UniversityINSERMParisFrance
| | - Elodie Segura
- Institut Curie, PSL Research UniversityINSERMParisFrance
| |
Collapse
|
3
|
Jani T, Santoro D, Shmalberg J. Investigation of the in vitro effects of cannabidiol, cannabidiolic acid, and the terpene β-caryophyllene on lymphocytes harvested from atopic and healthy dogs: A preliminary study. Res Vet Sci 2025; 182:105483. [PMID: 39616944 DOI: 10.1016/j.rvsc.2024.105483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
Cannabidiol (CBD) has been shown to have anti-inflammatory and antipruritic properties without the significant psychoactive effects. This study aims to evaluate the cytotoxic effects of, and the production of cytokines after exposure to CBD, cannabidiolic acid (CBDA), and β-caryophyllene (BCP), alone and in combination, by peripheral blood mononuclear cells (PBMC) from healthy and atopic dogs. Six healthy and five atopic, privately-owned dogs were enrolled. Peripheral blood mononuclear cells were harvested and incubated for 24 h with different concentrations of CBD, CBDA, and BCP alone or in combination. Cell viability and inflammatory cytokines were assessed. There was no difference in cell viability between baseline and tested concentrations of CBD, CBDA, or BCP in either healthy or in atopic PBMC. There was no effect of CBD, CBDA and BCP on the secretion of cytokines compared to baseline in healthy or atopic PMBC. The only exception was interleukin (IL)-10, increased in healthy PMBC exposed to CBD 100 ng/mL (p = 0.031) or CBDA 600 ng/mL (p = 0.017). Tumor necrosis factor (TNF)-α, monocyte chemoattractant protein (MCP-1), IL-2, and IL-18 were higher in atopic PBMC exposed to combinations of CBD, CBDA, and BCP compared to healthy post-exposure PBMC. This is the first study that tested the effect of CBD, CBDA, and BCP at different concentrations on atopic and healthy canine PBMC. The results of this study show that CBD, CBDA and BCP, at the tested concentrations, are safe for canine PBMC. However, CBD, CBDA and BCP did not show any direct anti-inflammatory effect under these experimental conditions. Further research is needed to confirm these results in a larger canine population.
Collapse
Affiliation(s)
- Twisha Jani
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, 2015 SW 16(th) Ave., Gainesville, FL 32610, USA
| | - Domenico Santoro
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, 2015 SW 16(th) Ave., Gainesville, FL 32610, USA.
| | - Justin Shmalberg
- Department of Comparative, Diagnostic and Population Medicine, College of Veterinary Medicine, University of Florida, 2015 SW 16th Avenue, Gainesville, FL 32610, USA
| |
Collapse
|
4
|
Anaya-Ayala JE, Galicia-Vega BJ, Mejia-Cervantes J, Ignacio-Alvarez E, Landero-Aguilar IA, Ferro-Flores G, Marquina-Castillo BN, Gibbens-Bandala B, Hinojosa CA. Analysis of small abdominal aortic aneurysms with the radiotracer technetium-99m- 6-hydrazinylnicotinoyl-C-C-chemokine receptor-2 ligand ( 99mTc-HYNIC-CCR2-L) with single-photon emission computed tomography. Pol J Radiol 2024; 89:e541-e548. [PMID: 39777328 PMCID: PMC11705040 DOI: 10.5114/pjr/194210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/07/2024] [Indexed: 01/11/2025] Open
Abstract
Purpose Monocyte chemoattractant protein-1 (MCP-1/CCL2) plays a key role for infiltration of monocytes/macrophages and studies have demonstrated that the MCP-1/C-C chemokine receptor 2 (CCR2) axis might be involved in the pathogenesis and progression of abdominal aortic aneurysms (AAA). Molecular imaging has shown potential for human clinical research studies. We evaluated the expression of CCR2 in patients with small AAA using single-photon emission computed tomography (SPECT) with the technetium-99m-6-hydrazinylnicotinoyl-C-C-chemokine receptor-2 ligand (99mTc-HYNIC-CCR2-L). Material and methods A pilot study was performed to evaluate patients with small asymptomatic AAA. The equipment used was a Symbia T2 (Siemens, Germany), with radiolabeled 99mTc-HYNIC-CCR2-L. The SPECT uptake and activity were assessed and counted based on the region of interest (ROI), with nonparametric statistics being employed to compare the aneurysms site, left ventricle (Control 1) and regions with a nondiseased aorta (Control 2). Results The three patients were male (100%) (mean age 81 years, and mean AAA maximum diameter of 40 mm, SD 3 mm). All patients tolerated the studies well. Images were obtained at one, two and four hours. The ROI mean value of the aneurysm site was 37,783 (SD 11,890), compared to the left ventricle (Control 1) 16,779 (SD 4397) (p-value = 0.0001); ROI for the nondiseased aortic region (Control 2) was significantly lower, 12,520 (SD 2141) (p-value = 0.0001). Conclusions Significant differences of CCR2 expression SPECT were found in the AAA site compared to the left ventricle and nondiseased aortic segments. The introduction of well-designed longitudinal studies with nuclear imaging modalities may assist in the molecular characterization of aneurysmal and rupture prediction.
Collapse
Affiliation(s)
- Javier E. Anaya-Ayala
- Department of Surgery, Section of Vascular Surgery and Endovascular Therapy, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Brenda J. Galicia-Vega
- Department of Nuclear Medicine, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Jacqueline Mejia-Cervantes
- Department of Surgery, Section of Vascular Surgery and Endovascular Therapy, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Eleazar Ignacio-Alvarez
- Department of Nuclear Medicine, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Ingrid A. Landero-Aguilar
- Department of Surgery, Section of Vascular Surgery and Endovascular Therapy, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Guillermina Ferro-Flores
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Mexico City, Mexico
| | - Brenda N. Marquina-Castillo
- Department of Pathology, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Brenda Gibbens-Bandala
- Department of Radioactive Materials, Instituto Nacional de Investigaciones Nucleares, Mexico City, Mexico
| | - Carlos A. Hinojosa
- Department of Surgery, Section of Vascular Surgery and Endovascular Therapy, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| |
Collapse
|
5
|
Domínguez-Luis MJ, Castro-Hernández J, Santos-Concepción S, Díaz-Martín A, Arce-Franco M, Pérez-González N, Díaz M, Castrillo A, Salido E, Machado JD, Gumá M, Corr M, Díaz-González F. Modulation of the K/BxN arthritis mouse model and the effector functions of human fibroblast-like synoviocytes by liver X receptors. Eur J Immunol 2024; 54:e2451136. [PMID: 39148175 DOI: 10.1002/eji.202451136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
The role of liver X receptors (LXR) in rheumatoid arthritis (RA) remains controversial. We studied the effect of LXR agonists on fibroblast-like synoviocytes (FLS) from RA patients and the K/BxN arthritis model in LXRα and β double-deficient (Nr1h2/3-/-) mice. Two synthetic LXR agonists, GW3965 and T0901317, were used to activate LXRs and investigate their effects on cell growth, proliferation and matrix metalloproteinases, and chemokine production in cultured FLS from RA patients. The murine model K/BxN serum transfer of inflammatory arthritis in Nr1h2/3-/- animals was used to investigate the role of LXRs on joint inflammation in vivo. LXR agonists inhibited the FLS proliferative capacity in response to TNF, the chemokine-induced migration, the collagenase activity in FLS supernatant and FLS CXCL12 production. In the K/BxN mouse model, Nr1h2/3-/- animals showed aggravated arthritis, histological inflammation, and joint destruction, as well as an increase in synovial metalloproteases and expression of proinflammatory mediators such as IL-1β and CCL2 in joints compared with wild type animals. Taken together, these data underscore the importance of LXRs in modulating the joint inflammatory response and highlight them as potential therapeutic targets in RA.
Collapse
MESH Headings
- Animals
- Humans
- Liver X Receptors/metabolism
- Liver X Receptors/genetics
- Mice
- Synoviocytes/metabolism
- Synoviocytes/pathology
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Fibroblasts/metabolism
- Mice, Knockout
- Disease Models, Animal
- Arthritis, Experimental/pathology
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Cells, Cultured
- Male
- Cell Proliferation
- Female
- Mice, Inbred C57BL
- Benzylamines/pharmacology
Collapse
Affiliation(s)
| | - Javier Castro-Hernández
- Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | | | - Ana Díaz-Martín
- Servicio de Reumatología, Hospital Universitario de Canarias, La Laguna, Spain
| | - Mayte Arce-Franco
- Servicio de Reumatología, Hospital Universitario de Canarias, La Laguna, Spain
| | | | - Mercedes Díaz
- Unidad de Biomedicina IIBM CSIC-Universidad de Las Palmas de Gran Canaria (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Las Palmas de Gran Canaria, Spain
| | - Antonio Castrillo
- Unidad de Biomedicina IIBM CSIC-Universidad de Las Palmas de Gran Canaria (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Las Palmas de Gran Canaria, Spain
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-Universidad Autónoma de Madrid, Madrid, Spain
| | - Eduardo Salido
- Departamento de Anatomía Patológica, Universidad de La Laguna, La Laguna, Spain
| | - José David Machado
- Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - Mónica Gumá
- Department of Medicine, University of California, San Diego, California, USA
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, California, USA
| | - Federico Díaz-González
- Servicio de Reumatología, Hospital Universitario de Canarias, La Laguna, Spain
- Departamento de Medicina Interna, Dermatología, Universidad de La Laguna, La Laguna, Spain
- Instituto Universitario de Tecnologías Biomédicas (ITB), Universidad de La Laguna, La Laguna, Spain
| |
Collapse
|
6
|
Pan SC, Wu YF, Lin YC, Cheng CM. Monocyte chemoattractant protein-1 detection in wound tissue fluids for the assisted diagnosis of wound infection. Surgery 2024; 176:154-161. [PMID: 38599982 DOI: 10.1016/j.surg.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Infections are commonly seen in wounds. The overall infection rate is 1.8% to 4.2%. Improper infection management can lead to serious conditions and may progress to life-threatening sepsis. Because there is a need for assistance in predicting wound infection before obvious clinical symptoms, the measurement of cytokines in wound tissue fluids has attracted our attention for determining the overall status of wound infection. Our intent was to assess the potential biomarkers in the diagnosis of wound infection. METHODS We collected 146 tissue fluids (acute: 59, chronic: 61, and normal: 26) for analysis of biomarkers using a human cytokine array. Serum C-reactive protein was also measured from 104 patients. The sensitivity and specificity of significant wound cytokines and serum C-reactive protein for the diagnosis of wound infection were evaluated. RESULTS Among biomarkers examined, serum C-reactive protein and tissue C-reactive protein were highly expressed in acute infection wounds, whereas monocyte chemoattractant protein-1 was significantly expressed in chronic infection wounds. Because the expression of wound biomarkers varied in different types of wounds, relationships among them were studied. A high correlation between tissue C-reactive protein and interleukin-8 (R2 = 0.7) and a moderate correlation between systemic and local C-reactive protein (R2 = 0.47) were observed. In addition, tissue monocyte chemoattractant protein-1 had better sensitivity (74%) and specificity (65%) in the diagnosis of wound infection. Moreover, combined serum C-reactive protein with monocyte chemoattractant protein-1 examination provided a higher area under the curve in the receiver operator characteristic curve (0.75). CONCLUSION We found that tissue monocyte chemoattractant protein-1 is a superior diagnostic marker for assistance with the diagnosis of wound infection.
Collapse
Affiliation(s)
- Shin-Chen Pan
- Section of Plastic and Reconstructive Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan.
| | - Yu-Feng Wu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsin-Chu, Taiwan; Section of Plastic and Reconstructive Surgery, Department of Surgery, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsin-Chu, Taiwan
| | - Yu-Chen Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsin-Chu, Taiwan
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsin-Chu, Taiwan
| |
Collapse
|
7
|
Zhang D, Wang M, Ma S, Liu M, Yu W, Zhang X, Liu T, Liu S, Ren X, Sun Q. Phosphoglycerate mutase 1 promotes breast cancer progression through inducing immunosuppressive M2 macrophages. Cancer Gene Ther 2024; 31:1018-1033. [PMID: 38750301 DOI: 10.1038/s41417-024-00769-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 07/20/2024]
Abstract
Immunosuppressive tumor microenvironment (TME) contributes to tumor progression and causes major obstacles for cancer therapy. Phosphoglycerate mutase 1 (PGAM1) is a key enzyme involved in cancer metabolism while its role in remodeling TME remains unclear. In this study, we reported that PGAM1 suppression in breast cancer (BC) cells led to a decrease in M2 polarization, migration, and interleukin-10 (IL-10) production of macrophages. PGAM1 regulation on CCL2 expression was essential to macrophage recruitment, which further mediated by activating JAK-STAT pathway. Additionally, the CCL2/CCR2 axis was observed to participate in PGAM1-mediated immunosuppression via regulating PD-1 expression in macrophages. Combined targeting of PGAM1 and the CCL2/CCR2 axis led to a reduction in tumor growth in vivo. Furthermore, clinical validation in BC tissues indicated a positive correlation between PGAM1, CCL2 and macrophage infiltration. Our study provides novel insights into the induction of immunosuppressive TME by PGAM1 and propose a new strategy for combination therapies targeting PGAM1 and macrophages in BC.
Collapse
Affiliation(s)
- Dong Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Min Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Shiya Ma
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Min Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenwen Yu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiying Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ting Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Shaochuan Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qian Sun
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
8
|
Ciechanowska A, Mika J. CC Chemokine Family Members' Modulation as a Novel Approach for Treating Central Nervous System and Peripheral Nervous System Injury-A Review of Clinical and Experimental Findings. Int J Mol Sci 2024; 25:3788. [PMID: 38612597 PMCID: PMC11011591 DOI: 10.3390/ijms25073788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Despite significant progress in modern medicine and pharmacology, damage to the nervous system with various etiologies still poses a challenge to doctors and scientists. Injuries lead to neuroimmunological changes in the central nervous system (CNS), which may result in both secondary damage and the development of tactile and thermal hypersensitivity. In our review, based on the analysis of many experimental and clinical studies, we indicate that the mechanisms occurring both at the level of the brain after direct damage and at the level of the spinal cord after peripheral nerve damage have a common immunological basis. This suggests that there are opportunities for similar pharmacological therapeutic interventions in the damage of various etiologies. Experimental data indicate that after CNS/PNS damage, the levels of 16 among the 28 CC-family chemokines, i.e., CCL1, CCL2, CCL3, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9, CCL11, CCL12, CCL17, CCL19, CCL20, CCL21, and CCL22, increase in the brain and/or spinal cord and have strong proinflammatory and/or pronociceptive effects. According to the available literature data, further investigation is still needed for understanding the role of the remaining chemokines, especially six of them which were found in humans but not in mice/rats, i.e., CCL13, CCL14, CCL15, CCL16, CCL18, and CCL23. Over the past several years, the results of studies in which available pharmacological tools were used indicated that blocking individual receptors, e.g., CCR1 (J113863 and BX513), CCR2 (RS504393, CCX872, INCB3344, and AZ889), CCR3 (SB328437), CCR4 (C021 and AZD-2098), and CCR5 (maraviroc, AZD-5672, and TAK-220), has beneficial effects after damage to both the CNS and PNS. Recently, experimental data have proved that blockades exerted by double antagonists CCR1/3 (UCB 35625) and CCR2/5 (cenicriviroc) have very good anti-inflammatory and antinociceptive effects. In addition, both single (J113863, RS504393, SB328437, C021, and maraviroc) and dual (cenicriviroc) chemokine receptor antagonists enhanced the analgesic effect of opioid drugs. This review will display the evidence that a multidirectional strategy based on the modulation of neuronal-glial-immune interactions can significantly improve the health of patients after CNS and PNS damage by changing the activity of chemokines belonging to the CC family. Moreover, in the case of pain, the combined administration of such antagonists with opioid drugs could reduce therapeutic doses and minimize the risk of complications.
Collapse
Affiliation(s)
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Str., 31-343 Kraków, Poland;
| |
Collapse
|
9
|
Massaro F, Corrillon F, Stamatopoulos B, Dubois N, Ruer A, Meuleman N, Bron D, Lagneaux L. Age-related changes in human bone marrow mesenchymal stromal cells: morphology, gene expression profile, immunomodulatory activity and miRNA expression. Front Immunol 2023; 14:1267550. [PMID: 38130717 PMCID: PMC10733451 DOI: 10.3389/fimmu.2023.1267550] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction Mesenchymal stromal cells (MSC) are one of the main cellular components of bone marrow (BM) microenvironment. MSC play a key role in tissue regeneration, but they are also capable of immunomodulating activity. With host aging, MSC undergo age-related changes, which alter these functions, contributing to the set-up of "inflammaging", which is known to be the basis for the development of several diseases of the elderly, including cancer. However, there's few data investigating this facet of MSC, mainly obtained using murine models or replicative senescence. The aim of this research was to identify morphological, molecular and functional alterations of human bone marrow-derived MSC from young (yBM-MSC) and old (oBM-MSC) healthy donors. Methods MSC were identified by analysis of cell-surface markers according to the ISCT criteria. To evaluate response to inflammatory status, MSC were incubated for 24h in the presence of IL-1β, IFN-α, IFN-ɣ and TNF-α. Macrophages were obtained by differentiation of THP-1 cells through PMA exposure. For M1 polarization experiments, a 24h incubation with LPS and IFN-ɣ was performed. MSC were plated at the bottom of the co-culture transwell system for all the time of cytokine exposure. Gene expression was evaluated by real-time PCR after RNA extraction from BM-MSC or THP-1 culture. Secreted cytokines levels were quantitated through ELISA assays. Results Aging MSC display changes in size, morphology and granularity. Higher levels of β-Gal, reactive oxygen species (ROS), IL-6 and IL-8 and impaired colony-forming and cell cycle progression abilities were found in oBM-MSC. Gene expression profile seems to vary according to subjects' age and particularly in oBM-MSC seem to be characterized by an impaired immunomodulating activity, with a reduced inhibition of macrophage M1 status. The comparative analysis of microRNA (miRNA) expression in yBM-MSC and oBM-MSC revealed a significant difference for miRNA known to be involved in macrophage polarization and particularly miR-193b-3p expression is strongly increased after co-culture of macrophages with yBM-MSC. Conclusion There are profound differences in terms of morphology, gene and miRNA expression and immunomodulating properties among yBM-MSC and oBM-MSC, supporting the critical role of aging BM microenvironment on senescence, immune-mediated disorders and cancer pathogenesis.
Collapse
Affiliation(s)
- Fulvio Massaro
- Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Florent Corrillon
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, ULB Cancer Research Center (U-CRC) - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Basile Stamatopoulos
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, ULB Cancer Research Center (U-CRC) - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nathan Dubois
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, ULB Cancer Research Center (U-CRC) - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Achille Ruer
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, ULB Cancer Research Center (U-CRC) - Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nathalie Meuleman
- Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Dominique Bron
- Department of Hematology, Jules Bordet Institute, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, ULB Cancer Research Center (U-CRC) - Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
10
|
Suenaga A, Seto Y, Funamoto M, Imanishi M, Tsuchiya K, Ikeda Y. TJ-17 (Goreisan) mitigates renal fibrosis in a mouse model of folic acid-induced chronic kidney disease. J Pharmacol Sci 2023; 153:31-37. [PMID: 37524452 DOI: 10.1016/j.jphs.2023.07.001] [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: 05/08/2023] [Revised: 06/22/2023] [Accepted: 07/04/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND AND PURPOSE TJ-17 (Goreisan), a traditional Japanese Kampo medicine, has been generally used to treat edema, such as heart failure, due to its diuretic effect. In the present study, we investigate the effects of TJ-17 on chronic kidney disease (CKD). METHODS We the preventive action of TJ-17 against acute kidney injury (AKI) transition to CKD in vivo using a folic acid (FA)-induced mouse model. Mice were treated with food containing TJ-17 at 48 h after FA intraperitoneal injection (AKI phase). RESULTS Histological analysis, as well as renal function and renal injury markers, deteriorated in mice with FA-induced CKD and were ameliorated by TJ-17 treatment. Increased levels of inflammatory cytokines and macrophage infiltration were also alleviated in mice treated with TJ-17. Renal fibrosis, a crucial factor in CKD, was induced by FA administration and inhibited by TJ-17 treatment. Pretreatment with TJ-17 did not exert an inhibitory effect on FA-induced AKI. The increase in urinary volume in FA-induced CKD mice was ameliorated by TJ-17 treatment, with a concurrent correction of reduced aquaporins expression in the kidney. CONCLUSION TJ-17 may have a novel preventive effect against inflammation, oxidative stress, and fibrosis, contributing to innovation in the treatment of CKD.
Collapse
Affiliation(s)
- Aoi Suenaga
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan; Student Lab, Faculty of Medicine, Tokushima University, Japan
| | - Yasuyuki Seto
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan; Student Lab, Faculty of Medicine, Tokushima University, Japan
| | - Masafumi Funamoto
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Masaki Imanishi
- Department of Medical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Koichiro Tsuchiya
- Department of Medical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yasumasa Ikeda
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| |
Collapse
|
11
|
Nagree MS, Rybova J, Kleynerman A, Ahrenhoerster CJ, Saville JT, Xu T, Bachochin M, McKillop WM, Lawlor MW, Pshezhetsky AV, Isaeva O, Budde MD, Fuller M, Medin JA. Spinal muscular atrophy-like phenotype in a mouse model of acid ceramidase deficiency. Commun Biol 2023; 6:560. [PMID: 37231125 DOI: 10.1038/s42003-023-04932-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/12/2023] [Indexed: 05/27/2023] Open
Abstract
Mutations in ASAH1 have been linked to two allegedly distinct disorders: Farber disease (FD) and spinal muscular atrophy with progressive myoclonic epilepsy (SMA-PME). We have previously reported FD-like phenotypes in mice harboring a single amino acid substitution in acid ceramidase (ACDase), P361R, known to be pathogenic in humans (P361R-Farber). Here we describe a mouse model with an SMA-PME-like phenotype (P361R-SMA). P361R-SMA mice live 2-3-times longer than P361R-Farber mice and have different phenotypes including progressive ataxia and bladder dysfunction, which suggests neurological dysfunction. We found profound demyelination, loss of axons, and altered sphingolipid levels in P361R-SMA spinal cords; severe pathology was restricted to the white matter. Our model can serve as a tool to study the pathological effects of ACDase deficiency on the central nervous system and to evaluate potential therapies for SMA-PME.
Collapse
Affiliation(s)
- Murtaza S Nagree
- Department of Medical Biophysics, University of Toronto, Toronto, M5G 1L7, ON, Canada
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Jitka Rybova
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Annie Kleynerman
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | | | - Jennifer T Saville
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, and Adelaide Medical School, University of Adelaide, Adelaide, SA, 5006, Australia
| | - TianMeng Xu
- CHU Sainte-Justine, Université de Montréal, Montréal, QC, H3T 1C5, Canada
| | | | - William M McKillop
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Michael W Lawlor
- Department of Pathology and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | | | - Olena Isaeva
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Matthew D Budde
- Clement J. Zablocki Veteran's Affairs Medical Center, Milwaukee, WI, 53295, USA
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Maria Fuller
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital, and Adelaide Medical School, University of Adelaide, Adelaide, SA, 5006, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Jeffrey A Medin
- Department of Medical Biophysics, University of Toronto, Toronto, M5G 1L7, ON, Canada.
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
12
|
Di Nardo A, Chang YL, Alimohammadi S, Masuda-Kuroki K, Wang Z, Sriram K, Insel PA. Mast cell tolerance in the skin microenvironment to commensal bacteria is controlled by fibroblasts. Cell Rep 2023; 42:112453. [PMID: 37120813 DOI: 10.1016/j.celrep.2023.112453] [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: 06/16/2022] [Revised: 01/16/2023] [Accepted: 04/14/2023] [Indexed: 05/01/2023] Open
Abstract
Activation and degranulation of mast cells (MCs) is an essential aspect of innate and adaptive immunity. Skin MCs, the most exposed to the external environment, are at risk of quickly degranulating with potentially severe consequences. Here, we define how MCs assume a tolerant phenotype via crosstalk with dermal fibroblasts (dFBs) and how this phenotype reduces unnecessary inflammation when in contact with beneficial commensal bacteria. We explore the interaction of human MCs (HMCs) and dFBs in the human skin microenvironment and test how this interaction controls MC inflammatory response by inhibiting the nuclear factor κB (NF-κB) pathway. We show that the extracellular matrix hyaluronic acid, as the activator of the regulatory zinc finger (de)ubiquitinating enzyme A20/tumor necrosis factor α-induced protein 3 (TNFAIP3), is responsible for the reduced HMC response to commensal bacteria. The role of hyaluronic acid as an anti-inflammatory ligand on MCs opens new avenues for the potential treatment of inflammatory and allergic disorders.
Collapse
Affiliation(s)
- Anna Di Nardo
- Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA.
| | - Yu-Ling Chang
- Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Shahrzad Alimohammadi
- Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Kana Masuda-Kuroki
- Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Zhenping Wang
- Department of Dermatology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Krishna Sriram
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA; Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| | - Paul A Insel
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA; Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
13
|
Qudus MS, Tian M, Sirajuddin S, Liu S, Afaq U, Wali M, Liu J, Pan P, Luo Z, Zhang Q, Yang G, Wan P, Li Y, Wu J. The roles of critical pro-inflammatory cytokines in the drive of cytokine storm during SARS-CoV-2 infection. J Med Virol 2023; 95:e28751. [PMID: 37185833 DOI: 10.1002/jmv.28751] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/17/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023]
Abstract
In patients with severe COVID-19, acute respiratory distress syndrome (ARDS), multiple organ dysfunction syndrome (MODS), and even mortality can result from cytokine storm, which is a hyperinflammatory medical condition caused by the excessive and uncontrolled release of pro-inflammatory cytokines. High levels of numerous crucial pro-inflammatory cytokines, such as interleukin-1 (IL-1), IL-2, IL-6, tumor necrosis factor-α, interferon (IFN)-γ, IFN-induced protein 10 kDa, granulocyte-macrophage colony-stimulating factor, monocyte chemoattractant protein-1, and IL-10 and so on, have been found in severe COVID-19. They participate in cascade amplification pathways of pro-inflammatory responses through complex inflammatory networks. Here, we review the involvements of these critical inflammatory cytokines in SARS-CoV-2 infection and discuss their potential roles in triggering or regulating cytokine storm, which can help to understand the pathogenesis of severe COVID-19. So far, there is rarely effective therapeutic strategy for patients with cytokine storm besides using glucocorticoids, which is proved to result in fatal side effects. Clarifying the roles of key involved cytokines in the complex inflammatory network of cytokine storm will help to develop an ideal therapeutic intervention, such as neutralizing antibody of certain cytokine or inhibitor of some inflammatory signal pathways.
Collapse
Affiliation(s)
- Muhammad Suhaib Qudus
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Mingfu Tian
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Summan Sirajuddin
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Siyu Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Uzair Afaq
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Muneeba Wali
- Department of Allied Health Sciences, CECOS University of IT and Emerging Sciences, Peshawar, Pakistan
| | - Jinbiao Liu
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Pan Pan
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Zhen Luo
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Qiwei Zhang
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Ge Yang
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Pin Wan
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Yongkui Li
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Foshan Institute of Medical Microbiology, Foshan, China
| |
Collapse
|
14
|
Ogura K, Endo M, Hase T, Negami H, Tsuchiya K, Nishiuchi T, Suzuki T, Ogai K, Sanada H, Okamoto S, Sugama J. Potential biomarker proteins for aspiration pneumonia detected by shotgun proteomics using buccal mucosa samples: a cross-sectional case-control study. Clin Proteomics 2023; 20:9. [PMID: 36894881 PMCID: PMC9996945 DOI: 10.1186/s12014-023-09398-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 02/21/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Aspiration pneumonia (AP), which is a major cause of death in the elderly, does present with typical symptoms in the early stages of onset, thus it is difficult to detect and treat at an early stage. In this study, we identified biomarkers that are useful for the detection of AP and focused on salivary proteins, which may be collected non-invasively. Because expectorating saliva is often difficult for elderly people, we collected salivary proteins from the buccal mucosa. METHODS We collected samples from the buccal mucosa of six patients with AP and six control patients (no AP) in an acute-care hospital. Following protein precipitation using trichloroacetic acid and washing with acetone, the samples were analyzed by liquid chromatography and tandem mass spectrometry (LC-MS/MS). We also determined the levels of cytokines and chemokines in non-precipitated samples from buccal mucosa. RESULTS Comparative quantitative analysis of LC-MS/MS spectra revealed 55 highly (P values < 0.10) abundant proteins with high FDR confidence (q values < 0.01) and high coverage (> 50%) in the AP group compared with the control group. Among the 55 proteins, the protein abundances of four proteins (protein S100-A7A, eukaryotic translation initiation factor 1, Serpin B4, and peptidoglycan recognition protein 1) in the AP group showed a negative correlation with the time post-onset; these proteins are promising AP biomarker candidates. In addition, the abundance of C-reactive protein (CRP) in oral samples was highly correlated with serum CRP levels, suggesting that oral CRP levels may be used as a surrogate to predict serum CRP in AP patients. A multiplex cytokine/chemokine assay revealed that MCP-1 tended to be low, indicating unresponsiveness of MCP-1 and its downstream immune pathways in AP. CONCLUSION Our findings suggest that oral salivary proteins, which are obtained non-invasively, can be utilized for the detection of AP.
Collapse
Affiliation(s)
- Kohei Ogura
- Advanced Health Care Science Research Unit, Institute for Frontier Science Initiative, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa, 9200942, Japan
| | - Maho Endo
- Advanced Health Care Science Research Unit, Institute for Frontier Science Initiative, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa, 9200942, Japan.,Nursing Department, Fujita Health University Hospital, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 4701192, Japan
| | - Takashi Hase
- Department of Oral and Maxillofacial Surgery, Noto General Hospital, 6-4 Fujibashi, Nanao, Ishikawa, 9260816, Japan
| | - Hitomi Negami
- Department of Oral and Maxillofacial Surgery, Noto General Hospital, 6-4 Fujibashi, Nanao, Ishikawa, 9260816, Japan
| | - Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University. Kakuma-Cho, Kanazawa, Ishikawa, 9201164, Japan
| | - Takumi Nishiuchi
- Division of Functional Genomics, Advanced Science Research Center, Kanazawa University, Kanazawa, Ishikawa, 9200934, Japan
| | - Takeshi Suzuki
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University. Kakuma-Cho, Kanazawa, Ishikawa, 9201164, Japan
| | - Kazuhiro Ogai
- Institute of Medical, Pharmaceutical and Health Sciences, AI Hospital/Macro Signal Dynamics Research and Development Center, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa, 9200942, Japan
| | - Hiromi Sanada
- Ishikawa Prefectural Nursing University, 1-1 Gakuendai, Kahoku, Ishikawa, 929-1210, Japan
| | - Shigefumi Okamoto
- Advanced Health Care Science Research Unit, Institute for Frontier Science Initiative, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa, 9200942, Japan. .,Department of Clinical Laboratory Sciences, Faculty of Health Sciences, Institute of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa, 9200942, Japan.
| | - Junko Sugama
- Research Center for Implementation Nursing Science Initiative, Innovation Promotion Division, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 4701192, Japan
| |
Collapse
|
15
|
Verçosa BLA, Muniz-Junqueira MI, Menezes-Souza D, Fujiwara RT, Borges LDF, Melo MN, Vasconcelos AC. MCP-1/IL-12 ratio expressions correlated with adventitial collagen depositions in renal vessels and IL-4/IFN-γ expression correlated with interstitial collagen depositions in the kidneys of dogs with canine leishmaniasis. Mol Immunol 2023; 156:61-76. [PMID: 36889187 DOI: 10.1016/j.molimm.2023.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/04/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023]
Abstract
Collagen deposition is a common event in chronic inflammation, and canine Leishmaniosis (CanL) is generally associated with a long and chronic evolution. Considering that the kidney shows fibrinogenic changes during CanL, and the balance of cytokines/chemokines regulates the profibrinogenic and antifibrinogenic immune responses differently, it can be hypothesized that the balance of cytokines/chemokines can be differentially expressed in the renal tissue in order to determine the expression of collagen depositions in the kidneys. This study aimed to measure collagen deposition and to evaluate cytokine/chemokine expressions in the kidney by means of qRT-PCR in sixteen Leishmania-infected dogs and six uninfected controls. Kidney fragments were stained with hematoxylin & eosin (H&E), Masson's Trichrome, Picrosirius Red, and Gomori's reticulin. Intertubular and adventitial collagen depositions were evaluated by the morphometric approach. Cytokine RNA expressions were measured by means of qRT-PCR to identify molecules involved in chronic collagen depositions in kidneys with CanL. Collagen depositions were related to the presence of clinical signs, and more intense intertubular collagen depositions occurred in infected dogs. Adventitial collagen deposition, as morphometrically measured by the average area of the collagen, was more intense in clinically affected dogs than in subclinically infected dogs. TNF-α/TGF-β, MCP1/IL-12, CCL5/IL-12, IL-4/IFN-γ, and IL-12/TGF-β expressions were associated with clinical manifestations in dogs with CanL. The IL-4/IFN-α ratio was more commonly expressed and upregulated in clinically affected dogs, and downregulated in subclinically infected dogs. Furthermore, MCP-1/IL-12 and CCL5/IL-12 were more commonly expressed in subclinically infected dogs. Strong positive correlations were detected between morphometric values of interstitial collagen depositions and MCP-1/IL-12, IL-12, and IL-4 mRNA expression levels in the renal tissues. Adventitial collagen deposition was correlated with TGF-β, IL-4/IFN-γ, and TNF-α/TGF-β. In conclusion, our results showed the association of MCP-1/IL-12 and CCL5/IL-12 ratios with an absence of clinical signs, as well as an IL-4/IFN-α ratio with adventitial and intertubular collagen depositions in dogs with visceral leishmaniosis.
Collapse
Affiliation(s)
- Barbara Laurice Araújo Verçosa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Imunologia Celular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil.
| | | | - Daniel Menezes-Souza
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo Toshio Fujiwara
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luciano de F Borges
- Instituto de Ciências Biológicas, Universidade Federal de São Paulo, São Paulo, São Paulo, Brazil
| | - Maria Norma Melo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anilton Cesar Vasconcelos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
16
|
Hsu RJ, Yu WC, Peng GR, Ye CH, Hu S, Chong PCT, Yap KY, Lee JYC, Lin WC, Yu SH. The Role of Cytokines and Chemokines in Severe Acute Respiratory Syndrome Coronavirus 2 Infections. Front Immunol 2022; 13:832394. [PMID: 35464491 PMCID: PMC9021400 DOI: 10.3389/fimmu.2022.832394] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/24/2022] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in countless infections and caused millions of deaths since its emergence in 2019. Coronavirus disease 2019 (COVID-19)-associated mortality is caused by uncontrolled inflammation, aberrant immune response, cytokine storm, and an imbalanced hyperactive immune system. The cytokine storm further results in multiple organ failure and lung immunopathology. Therefore, any potential treatments should focus on the direct elimination of viral particles, prevention strategies, and mitigation of the imbalanced (hyperactive) immune system. This review focuses on cytokine secretions of innate and adaptive immune responses against COVID-19, including interleukins, interferons, tumor necrosis factor-alpha, and other chemokines. In addition to the review focus, we discuss potential immunotherapeutic approaches based on relevant pathophysiological features, the systemic immune response against SARS-CoV-2, and data from recent clinical trials and experiments on the COVID-19-associated cytokine storm. Prompt use of these cytokines as diagnostic markers and aggressive prevention and management of the cytokine storm can help determine COVID-19-associated morbidity and mortality. The prophylaxis and rapid management of the cytokine storm appear to significantly improve disease outcomes. For these reasons, this study aims to provide advanced information to facilitate innovative strategies to survive in the COVID-19 pandemic.
Collapse
Affiliation(s)
- Ren-Jun Hsu
- Cancer Center, Hualien Tzu Chi Hospital, Buddhist Tzuchi Medical Foundation, Hualien, Taiwan.,School of Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wei-Chieh Yu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Guan-Ru Peng
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Chih-Hung Ye
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - SuiYun Hu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | | | - Kah Yi Yap
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | | | - Wei-Chen Lin
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Shu-Han Yu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
17
|
Wang B, Zhang X, Chen H, Koh A, Zhao C, Chen Y. A Review of Intraocular Biomolecules in Retinal Vein Occlusion: Toward Potential Biomarkers for Companion Diagnostics. Front Pharmacol 2022; 13:859951. [PMID: 35559255 PMCID: PMC9086509 DOI: 10.3389/fphar.2022.859951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/25/2022] [Indexed: 12/04/2022] Open
Abstract
Retinal vein occlusion (RVO) is one of the most common retinal vascular diseases. The pathogenesis of RVO is multifactorial and involves a complex interplay among a variety of vascular and inflammatory mediators. Many cytokines, chemokines, growth factors, and cell adhesion molecules have been reported to be implicated. Treatments for RVO are directed at the management of underlying risk factors and vision-threatening complications, including macula edema (ME) and neovascularization. Intravitreal anti-VEGF agents are currently considered as the first-line treatment for ME secondary to RVO (RVO-ME), but a substantial proportion of patients responded insufficiently to anti-VEGF agents. Since RVO-ME refractory to anti-VEGF agents generally responds to corticosteroids and its visual outcome is negatively correlated to disease duration, prediction of treatment response at baseline in RVO-ME may significantly improve both cost-effectiveness and visual prognosis. Several bioactive molecules in the aqueous humor were found to be associated with disease status in RVO. This review aims to present a comprehensive review of intraocular biomolecules reported in RVO, including VEGF, IL-6, IL-8, MCP-1, sICAM-1, IL-12, IL-13, sVEGFR-1, sVEGFR-2, PDGF-AA, etc., highlighting their association with disease severity and/or phenotype, and their potential roles in prognostic prediction and treatment selection. Some of these molecules may serve as biomarkers for aqueous humor-based companion diagnostics for the treatment of RVO in the future.
Collapse
Affiliation(s)
- Bingjie Wang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Xiao Zhang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huan Chen
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Adrian Koh
- Eye & Retina Surgeons, Camden Medical Centre, Singapore, Singapore
| | - Chan Zhao
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Youxin Chen
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Nording H, Sauter M, Lin C, Steubing R, Geisler S, Sun Y, Niethammer J, Emschermann F, Wang Y, Zieger B, Nieswandt B, Kleinschnitz C, Simon DI, Langer HF. Activated Platelets Upregulate β 2 Integrin Mac-1 (CD11b/CD18) on Dendritic Cells, Which Mediates Heterotypic Cell-Cell Interaction. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1729-1741. [PMID: 35277420 DOI: 10.4049/jimmunol.2100557] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 01/11/2022] [Indexed: 12/30/2022]
Abstract
Recent evidence suggests interaction of platelets with dendritic cells (DCs), while the molecular mechanisms mediating this heterotypic cell cross-talk are largely unknown. We evaluated the role of integrin Mac-1 (αMβ2, CD11b/CD18) on DCs as a counterreceptor for platelet glycoprotein (GP) Ibα. In a dynamic coincubation model, we observed interaction of human platelets with monocyte-derived DCs, but also that platelet activation induced a sharp increase in heterotypic cell binding. Inhibition of CD11b or GPIbα led to significant reduction of DC adhesion to platelets in vitro independent of GPIIbIIIa, which we confirmed using platelets from Glanzmann thrombasthenia patients and transgenic mouse lines on C57BL/6 background (GPIbα-/-, IL4R-GPIbα-tg, and muMac1 mice). In vivo, inhibition or genetic deletion of CD11b and GPIbα induced a significant reduction of platelet-mediated DC adhesion to the injured arterial wall. Interestingly, only intravascular antiCD11b inhibited DC recruitment, suggesting a dynamic DC-platelet interaction. Indeed, we could show that activated platelets induced CD11b upregulation on Mg2+-preactivated DCs, which was related to protein kinase B (Akt) and dependent on P-selectin and P-selectin glycoprotein ligand 1. Importantly, specific pharmacological targeting of the GPIbα-Mac-1 interaction site blocked DC-platelet interaction in vitro and in vivo. These results demonstrate that cross-talk of platelets with DCs is mediated by GPIbα and Mac-1, which is upregulated on DCs by activated platelets in a P-selectin glycoprotein ligand 1-dependent manner.
Collapse
Affiliation(s)
- Henry Nording
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany.,German Research Centre for Cardiovascular Research, Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany.,University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Manuela Sauter
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany.,University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Chaolan Lin
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Rebecca Steubing
- Department of Neurology and Center for Translational and Behavioral Neurosciences, University Hospital Essen, Essen, Germany
| | - Sven Geisler
- Cell Analysis Core Facility, University of Lübeck, Lübeck, Germany
| | - Ying Sun
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Joel Niethammer
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Fréderic Emschermann
- Department of Cardiovascular Medicine, University Hospital, Eberhard Karls University, Tübingen, Germany
| | - Yunmei Wang
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine and Harrington Heart & Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Barbara Zieger
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany; and
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational and Behavioral Neurosciences, University Hospital Essen, Essen, Germany
| | - Daniel I Simon
- Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine and Harrington Heart & Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH.,University Hospitals Cleveland Medical Center, Cleveland, OH
| | - Harald F Langer
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany; .,German Research Centre for Cardiovascular Research, Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany.,University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| |
Collapse
|
19
|
Zhao D, Zhang X, Feng Y, Bian Y, Fu Z, Wu Y, Ma Y, Li C, Wang J, Dai J, Fu Y, Lin S, Hu J. Taurine Alleviates LPS-Induced Acute Lung Injury by Suppressing TLR-4/NF-κB Pathway. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1370:63-72. [DOI: 10.1007/978-3-030-93337-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
20
|
Tilstam PV, Schulte W, Holowka T, Kim BS, Nouws J, Sauler M, Piecychna M, Pantouris G, Lolis E, Leng L, Bernhagen J, Fingerle-Rowson G, Bucala R. MIF but not MIF-2 recruits inflammatory macrophages in an experimental polymicrobial sepsis model. J Clin Invest 2021; 131:127171. [PMID: 34850744 DOI: 10.1172/jci127171] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 10/14/2021] [Indexed: 11/17/2022] Open
Abstract
Excessive inflammation drives the progression from sepsis to septic shock. Macrophage migration inhibitory factor (MIF) is of interest because MIF promoter polymorphisms predict mortality in different infections, and anti-MIF antibody improves survival in experimental models when administered 8 hours after infectious insult. The recent description of a second MIF superfamily member, D-dopachrome tautomerase (D-DT/MIF-2), prompted closer investigation of MIF-dependent responses. We subjected Mif-/- and Mif-2-/- mice to polymicrobial sepsis and observed a survival benefit with Mif but not Mif-2 deficiency. Survival was associated with reduced numbers of small peritoneal macrophages (SPMs) that, in contrast to large peritoneal macrophages (LPMs), were recruited into the peritoneal cavity. LPMs produced higher quantities of MIF than SPMs, but SPMs expressed higher levels of inflammatory cytokines and the MIF receptors CD74 and CXCR2. Adoptive transfer of WT SPMs into Mif-/- hosts reduced the protective effect of Mif deficiency in polymicrobial sepsis. Notably, MIF-2 lacks the pseudo-(E)LR motif present in MIF that mediates CXCR2 engagement and SPM migration, supporting a specific role for MIF in the recruitment and accumulation of inflammatory SPMs.
Collapse
Affiliation(s)
- Pathricia Veronica Tilstam
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| | - Wibke Schulte
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Charité, Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Thomas Holowka
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Bong-Sung Kim
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Plastic, Reconstructive and Hand Surgery, RWTH Aachen University, Aachen, Germany.,Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Jessica Nouws
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Maor Sauler
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marta Piecychna
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Georgios Pantouris
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Chemistry, University of the Pacific, Stockton, California, USA
| | - Elias Lolis
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jürgen Bernhagen
- Department of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig-Maximilians-University Munich, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| | - Günter Fingerle-Rowson
- Department I of Internal Medicine, University of Cologne, Center for Integrated Oncology Aachen Bonn Köln Düsseldorf, Cologne, Germany
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
21
|
Mohyuddin SG, Qamar A, Hu CY, Li Y, Chen SW, Wen JY, Bao ML, Ju XH. Terpinen4-ol inhibits heat stress induced inflammation in colonic tissue by Activating Occludin, Claudin-2 and TLR4/NF-κB signaling pathway. Int Immunopharmacol 2021; 99:107727. [PMID: 34426115 DOI: 10.1016/j.intimp.2021.107727] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/13/2021] [Accepted: 04/25/2021] [Indexed: 01/18/2023]
Abstract
Heat stress has severe implications on the health of mice involving intestinal mucosal barrier damage and dysregulated mucosal immune response. This study was designed with long-term heat stress to detect the protective effect of terpinen4-ol on body weight, colon length, organ index, morphological structure, inflammatory cytokines expression, Claudin-2, Occludin, and TLR4 signaling pathway of colonic tissue in mice under heat stress. A study found that oral administration of terpinen4-ol helped against mortality and intestinal inflammation in a mouse model of acute colitis induced by heat stress (40 °C per day for 4 h) exposed for 14 consecutive days. The mice were divided into five groups including control, heat stress, terpinen4-ol low dose (TER LD: 5 mg/kg), medium dose (TER MD: 10 mg/kg), and high dose (TER HD: 20 mg/kg) group. Our study showed that the heat-stress terpinen4-ol group had improved body weight, colon length, and organ index, the number of white blood cells, lymphocytes, and neutrophils in the blood as compared to the heat stress group. In addition, results showed that heat stress upregulated the expression of TLR4, p65, TNF-α, and IL-10. While, in mice receiving the oral administration of terpinen4-ol, the production of TNF-α, IL-10, TLR4, and p65 was suppressed on day 1, 7, and 14 of heat stress. In addition Claudin-2, Occludin mRNA levels were upregulated in mice receiving terpinen4-ol on day 1, 7, and 14 of heat stress. Furthermore, the IL-6, IL-10, TNF-α serum levels were also upregulated in mice under heat stress, but in mice receiving the oral administration of terpinen4-ol, the IL-6, IL-10, TNF-α level was down-regulated on day 1, 7, and 14 of heat stress. Histomorphological examination found that as compared to the control group, the muscle layer thickness and villi height of mice in the heat stress group were significantly reduced, while the changes of the above indicators in the terpinene4-ol groups were improved than those in the heat stress group. In conclusion, the terpinen4-ol has a protective effect on colonic tissue damage induced by heat stress.
Collapse
Affiliation(s)
- Sahar Ghulam Mohyuddin
- Department of Animal Science, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524088, China
| | - Aftab Qamar
- Department of Animal Science, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524088, China
| | - Can-Ying Hu
- Department of Animal Science, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524088, China
| | - Yun Li
- Department of Animal Science, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524088, China
| | - Sheng-Wei Chen
- Department of Veterinary Medicine, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524088, China
| | - Jia-Ying Wen
- Department of Veterinary Medicine, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524088, China
| | - Ming-Long Bao
- Department of Veterinary Medicine, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524088, China
| | - Xiang Hong Ju
- Department of Veterinary Medicine, College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang, Guangdong 524088, China.
| |
Collapse
|
22
|
The Evolving Roles of Cardiac Macrophages in Homeostasis, Regeneration, and Repair. Int J Mol Sci 2021; 22:ijms22157923. [PMID: 34360689 PMCID: PMC8347787 DOI: 10.3390/ijms22157923] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Macrophages were first described as phagocytic immune cells responsible for maintaining tissue homeostasis by the removal of pathogens that disturb normal function. Historically, macrophages have been viewed as terminally differentiated monocyte-derived cells that originated through hematopoiesis and infiltrated multiple tissues in the presence of inflammation or during turnover in normal homeostasis. However, improved cell detection and fate-mapping strategies have elucidated the various lineages of tissue-resident macrophages, which can derive from embryonic origins independent of hematopoiesis and monocyte infiltration. The role of resident macrophages in organs such as the skin, liver, and the lungs have been well characterized, revealing functions well beyond a pure phagocytic and immunological role. In the heart, recent research has begun to decipher the functional roles of various tissue-resident macrophage populations through fate mapping and genetic depletion studies. Several of these studies have elucidated the novel and unexpected roles of cardiac-resident macrophages in homeostasis, including maintaining mitochondrial function, facilitating cardiac conduction, coronary development, and lymphangiogenesis, among others. Additionally, following cardiac injury, cardiac-resident macrophages adopt diverse functions such as the clearance of necrotic and apoptotic cells and debris, a reduction in the inflammatory monocyte infiltration, promotion of angiogenesis, amelioration of inflammation, and hypertrophy in the remaining myocardium, overall limiting damage extension. The present review discusses the origin, development, characterization, and function of cardiac macrophages in homeostasis, cardiac regeneration, and after cardiac injury or stress.
Collapse
|
23
|
NLRP3-Inflammasome Inhibition during Respiratory Virus Infection Abrogates Lung Immunopathology and Long-Term Airway Disease Development. Viruses 2021; 13:v13040692. [PMID: 33923693 PMCID: PMC8072578 DOI: 10.3390/v13040692] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/16/2022] Open
Abstract
Respiratory syncytial virus (RSV) infects most infants by two years of age. It can cause severe disease leading to an increased risk of developing asthma later in life. Previously, our group has shown that RSV infection in mice and infants promotes IL-1β production. Here, we characterized the role of NLRP3-Inflammasome activation during RSV infection in adult mice and neonates. We observed that the inhibition of NLRP3 activation using the small molecule inhibitor, MCC950, or in genetically modified NLRP3 knockout (Nlrp3−/−) mice during in vivo RSV infection led to decreased lung immunopathology along with a reduced expression of the mucus-associated genes and reduced production of innate cytokines (IL-1β, IL-33 and CCL2) linked to severe RSV disease while leading to significant increases in IFN-β. NLRP3-inflammasome inhibition or deletion diminished Th2 cytokines and inflammatory cell infiltration into the lungs. Furthermore, NLRP3 inhibition or deletion during early-life RSV infection led to reducing viral-exacerbated allergic response in a mouse model of RSV-induced allergy exacerbation. Here, we demonstrated the critical role of NLRP3-inflammasome activation in RSV immunopathology and the related long-term airway alteration. Moreover, these findings suggest the NLRP3-inflammasome as a potential therapeutic target to attenuate severe RSV disease and limit childhood asthma development.
Collapse
|
24
|
Moadab F, Khorramdelazad H, Abbasifard M. Role of CCL2/CCR2 axis in the immunopathogenesis of rheumatoid arthritis: Latest evidence and therapeutic approaches. Life Sci 2021; 269:119034. [PMID: 33453247 DOI: 10.1016/j.lfs.2021.119034] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/02/2021] [Accepted: 01/07/2021] [Indexed: 12/18/2022]
Abstract
Evidence suggests that uncontrolled immune system responses and their components play a significant role in developing rheumatoid arthritis (RA), which is considered an autoimmune disease (AD). Among immune system mediators, cytokines and chemokines are involved in numerous physiological and pathological processes. CCL2 or monocyte chemoattractant protein-1 (MCP-1) is known as a CC chemokine that can induce the locomotion and recruitment of monocytes and macrophages to the site of injury. When CCL2 binds to its receptors, the most important of which is CCR2, various signaling pathways are triggered, eventually leading to various immunological events such as inflammation. This chemokine also participates in several events involved in RA pathogenesis, such as osteoclastogenesis, migration of effector T cells to the RA synovium tissue, and angiogenesis. In this review article, the role of the CCL2/CCR2 axis in RA pathogenesis and the immunotherapy opportunities based on CCL2/CCR2 axis targeting has been discussed based on existing investigations.
Collapse
Affiliation(s)
- Fatemeh Moadab
- Student Research Committee, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Immunology, School of Medicine; Molecular Medicine Research Center, Institute of Basic Medical Sciences Research, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine; Molecular Medicine Research Center, Institute of Basic Medical Sciences Research, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mitra Abbasifard
- Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine; Molecular Medicine Research Center, Institute of Basic Medical Sciences Research, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
25
|
Popiolek-Barczyk K, Ciechanowska A, Ciapała K, Pawlik K, Oggioni M, Mercurio D, De Simoni MG, Mika J. The CCL2/CCL7/CCL12/CCR2 pathway is substantially and persistently upregulated in mice after traumatic brain injury, and CCL2 modulates the complement system in microglia. Mol Cell Probes 2020; 54:101671. [PMID: 33160071 DOI: 10.1016/j.mcp.2020.101671] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/15/2020] [Accepted: 11/01/2020] [Indexed: 12/28/2022]
Abstract
Traumatic brain injury (TBI) is the leading cause of death in the global population. Disturbed inflammatory processes after TBI exacerbate secondary brain injury and contribute to unfavorable outcomes. Multiple inflammatory events that accompany brain trauma, such as glial activation, chemokine release, or the initiation of the complement system cascade, have been identified as potential targets for TBI treatment. However, the participation of chemokines in the complement activation remains unknown. Our studies sought to determine the changes in the expression of the molecules involved in the CCL2/CCL7/CCL12/CCR2 pathway in the injured brain and the effect of CCL2, CCL7, and CCL12 (10, 100, and 500 ng/mL) on the classic and lectin complement pathways and inflammatory factors in microglial cell cultures. Brain injury in mice was modeled by controlled cortical impact (CCI). Our findings indicate a time-dependent upregulation of CCL2, CCL7, and CCL12 at the mRNA and protein levels within the cortex, striatum, and/or thalamus beginning 24 h after the trauma. The analysis of the expression of the receptor of the tested chemokines, CCR2, revealed its substantial upregulation within the injured brain areas mainly on the mRNA level. Using primary cortical microglial cell cultures, we observed a substantial increase in the expression of CCL2, CCL7, and CCL12 after 24 h of LPS (100 ng/mL) treatment. CCL2 stimulation of microglia increased the level of IL-1β mRNA but did not influence the expression of IL-18, IL-6, and IL-10. Moreover, CCL2 significantly increased the expression of Iba1, a marker of microglia activation. CCL2 and CCL12 upregulated the expression of C1qa but did not influence the expression of C1ra and C1s1 (classical pathway); moreover, CCL2 increased ficolin A expression and reduced collectin 11 expression (lectin pathway). Additionally, we observed the downregulation of pentraxin 3, a modulator of the complement cascade, after CCL2 and CCL12 treatment. We did not detect the expression of ficolin B, Mbl1, and Mbl2 in microglial cells. Our data identify CCL2 as a modulator of the classical and lectin complement pathways suggesting that CCL2 may be a promising target for pharmacological intervention after brain injury. Moreover, our study provides evidence that CCL2 and two other CCR2 ligands may play a role in the development of changes in TBI.
Collapse
Affiliation(s)
- Katarzyna Popiolek-Barczyk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Agata Ciechanowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Katarzyna Ciapała
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Katarzyna Pawlik
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Marco Oggioni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Domenico Mercurio
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Maria-Grazia De Simoni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Joanna Mika
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland.
| |
Collapse
|
26
|
Dewhurst MR, Ow JR, Zafer G, van Hul NKM, Wollmann H, Bisteau X, Brough D, Choi H, Kaldis P. Loss of hepatocyte cell division leads to liver inflammation and fibrosis. PLoS Genet 2020; 16:e1009084. [PMID: 33147210 PMCID: PMC7641358 DOI: 10.1371/journal.pgen.1009084] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
The liver possesses a remarkable regenerative capacity based partly on the ability of hepatocytes to re-enter the cell cycle and divide to replace damaged cells. This capability is substantially reduced upon chronic damage, but it is not clear if this is a cause or consequence of liver disease. Here, we investigate whether blocking hepatocyte division using two different mouse models affects physiology as well as clinical liver manifestations like fibrosis and inflammation. We find that in P14 Cdk1Liv-/- mice, where the division of hepatocytes is abolished, polyploidy, DNA damage, and increased p53 signaling are prevalent. Cdk1Liv-/- mice display classical markers of liver damage two weeks after birth, including elevated ALT, ALP, and bilirubin levels, despite the lack of exogenous liver injury. Inflammation was further studied using cytokine arrays, unveiling elevated levels of CCL2, TIMP1, CXCL10, and IL1-Rn in Cdk1Liv-/- liver, which resulted in increased numbers of monocytes. Ablation of CDK2-dependent DNA re-replication and polyploidy in Cdk1Liv-/- mice reversed most of these phenotypes. Overall, our data indicate that blocking hepatocyte division induces biological processes driving the onset of the disease phenotype. It suggests that the decrease in hepatocyte division observed in liver disease may not only be a consequence of fibrosis and inflammation, but also a pathological cue.
Collapse
Affiliation(s)
- Matthew R. Dewhurst
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
- Lydia Becker Institute of Immunology and Inflammation; and Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Jin Rong Ow
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Gözde Zafer
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
- Department of Biochemistry, National University of Singapore (NUS), Singapore
| | - Noémi K. M. van Hul
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Heike Wollmann
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Xavier Bisteau
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - David Brough
- Lydia Becker Institute of Immunology and Inflammation; and Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Hyungwon Choi
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
- Department of Biochemistry, National University of Singapore (NUS), Singapore
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Sweden
- * E-mail:
| |
Collapse
|
27
|
Vaccination with Secreted Aspartyl Proteinase 2 Protein from Candida parapsilosis Can Enhance Survival of Mice during C. tropicalis-Mediated Systemic Candidiasis. Infect Immun 2020; 88:IAI.00312-20. [PMID: 32661125 DOI: 10.1128/iai.00312-20] [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: 05/27/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022] Open
Abstract
The rising incidence of non-albicans Candida species globally, along with the emergence of drug resistance, is a cause for concern. This study investigated the protective efficacy of secreted aspartyl proteinase 2 (Sap2) in systemic C. tropicalis infection. Vaccination with recombinant Sap2 (rSap2) protein from C. parapsilosis enhanced survival of mice compared to rSap2 vaccinations from C. albicans (P = 0.02), C. tropicalis (P = 0.06), and sham immunization (P = 0.04). Compared to sham-immunized mice, the fungal CFU number was significantly reduced in organs of Sap2-parapsilosis-immunized mice. Histopathologically, increased neutrophilic recruitment was observed in Sap2-parapsilosis- and Sap2-tropicalis-immunized mice. Among different rSap2 proteins, Sap2-parapsilosis vaccination induced increased titers of Sap2-specific Ig, IgG, and IgM antibodies, which could bind whole fungus. Between different groups, sera from Sap2-parapsilosis-vaccinated mice exhibited increased C. tropicalis biofilm inhibition ability in vitro and enhanced neutrophil-mediated fungal killing. Passive transfer of anti-Sap2-parapsilosis immune serum in naive mice significantly reduced fungal burdens compared to those in mice receiving anti-sham immune serum. Higher numbers of plasma cells and Candida-binding B cells in Sap2-vaccinated mice suggest a role of B cells during early stages of Sap2-mediated immune response. Additionally, increased levels of Th1/Th2/Th17 cytokines observed in Sap2-parapsilosis-vaccinated mice indicate immunomodulatory properties of Sap2. Epitope analysis performed using identified B-cell epitopes provides a basis to understand differences in immunogenicity observed among Sap2-antigens and can aid the development of a multivalent or multiepitope anti-Candida vaccine(s). In summary, our results suggest that Sap2-parapsilosis vaccination can improve mouse survival during C. tropicalis infection by inducing both humoral and cellular immunity, and higher titers of Sap2-induced antibodies are beneficial during systemic candidiasis.
Collapse
|
28
|
Crosse EI, Gordon-Keylock S, Rybtsov S, Binagui-Casas A, Felchle H, Nnadi NC, Kirschner K, Chandra T, Tamagno S, Webb DJ, Rossi F, Anderson RA, Medvinsky A. Multi-layered Spatial Transcriptomics Identify Secretory Factors Promoting Human Hematopoietic Stem Cell Development. Cell Stem Cell 2020; 27:822-839.e8. [PMID: 32946788 PMCID: PMC7671940 DOI: 10.1016/j.stem.2020.08.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/25/2020] [Accepted: 08/07/2020] [Indexed: 01/30/2023]
Abstract
Hematopoietic stem cells (HSCs) first emerge in the embryonic aorta-gonad-mesonephros (AGM) region. Studies of model organisms defined intersecting signaling pathways that converge to promote HSC emergence predominantly in the ventral domain of the dorsal aorta. Much less is known about mechanisms driving HSC development in humans. Here, to identify secreted signals underlying human HSC development, we combined spatial transcriptomics analysis of dorsoventral polarized signaling in the aorta with gene expression profiling of sorted cell populations and single cells. Our analysis revealed a subset of aortic endothelial cells with a downregulated arterial signature and a predicted lineage relationship with the emerging HSC/progenitor population. Analysis of the ventrally polarized molecular landscape identified endothelin 1 as an important secreted regulator of human HSC development. The obtained gene expression datasets will inform future studies on mechanisms of HSC development in vivo and on generation of clinically relevant HSCs in vitro.
Spatial transcriptome profiling of the human HSC developmental niche Characterization of an HSC precursor population at single-cell resolution Cardiac EGF pathway is ventrally enriched next to developing IAHCs/HSCs Ventrally secreted endothelin promotes development of HSCs
Collapse
Affiliation(s)
- Edie I Crosse
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | | | - Stanislav Rybtsov
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Anahi Binagui-Casas
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Hannah Felchle
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Nneka C Nnadi
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Kristina Kirschner
- Institute of Cancer Sciences, University of Glasgow, Bearsden G61 1QH, UK
| | - Tamir Chandra
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Sara Tamagno
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - David J Webb
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Fiona Rossi
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh EH16 4TJ UK
| | - Alexander Medvinsky
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK.
| |
Collapse
|
29
|
Li H, Chen Z, Li J, Liu R, Zhao F, Liu R. Indium oxide nanoparticles induce lung intercellular toxicity between bronchial epithelial cells and macrophages. J Appl Toxicol 2020; 40:1636-1646. [PMID: 32608070 DOI: 10.1002/jat.4023] [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] [Received: 01/14/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
Concerns have been raised over the safety and health of industrial workers exposed to indium oxide nanoparticles (IO-NPs) when working. IO-NPs were previously shown in vitro and in vivo to be cytotoxic, but the mechanism of pathogenesis was unclear. In this study, the effects of IO-NPs on lung cells associated with respiratory and immune barriers and the toxic effects of intercellular cascades were studied. Here IO-NPs had acute toxicity to Wistar rats over a time course (5 days post-intratracheal instillation). Following treatment epithelial cells (16HBE) or macrophages (RAW264.7) with IO-NPs or IO fine particles (IO-FPs), the damage of 16HBE cells caused by IO-NPs was serious, mainly in the mitochondrial and rough endoplasmic reticulum. The lactate dehydrogenase level also showed that cytotoxicity in vitro was more serious for IO-NPs compared with IO-FPs. The level of In3+ (examined by inductively coupled plasma mass spectrometry) in 16HBE cells was 10 times higher than that in RAW cells. In3+ , releasing from IO-NPs absorbed by 16HBE cells, could not only significantly inhibit the phagocytosis and migration of macrophages (P < .0001), but also stimulate RAW cells to secrete high levels of inflammatory cytokines. IO-NPs can directly damage pulmonary epithelial cells. The In3+ released by epithelial cells affect the phagocytosis and migration of macrophages, which may be a new point for the decrease in the clearance of alveolar surfactants and the development of IO-related pulmonary alveolar proteinosis.
Collapse
Affiliation(s)
- Huilin Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Zhaofang Chen
- Shool of the Environment, Nanjing University, Nanjing, China
| | - Jinxia Li
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing, China
| | - Ru Liu
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing, China
| | - Feng Zhao
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Science, Beijing, China
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
30
|
Chen Z, Yin S, Zheng L, Tang W, Kang M, Wei W, Sui K. Relationship between the Monocyte Chemo-attractant Protein-1 gene rs1024611 A>G Polymorphism and Cancer Susceptibility: A Meta-analysis Involving 14,617 Subjects. Immunol Invest 2020; 50:461-477. [PMID: 32552226 DOI: 10.1080/08820139.2020.1776726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Inflammatory and inducible chemokines are the hallmarks of malignancy. Monocyte chemo-attractant protein-1 (MCP-1) is a crucial chemokine implicated in infection and inflammation. Methods: We performed an updated meta-analysis of thirty independent case-control studies with 6,777 cancer cases and 7,840 controls to determine if the MCP-1 gene rs1024611 A > G variant is associated with the risk of cancer. Results: The G allele carriers of rs1024611 in the MCP-1 gene might have a null association with cancer risk in overall comparison. In a subgroup analysis by ethnicity, we identified a marked association between the MCP-1 G allele rs1024611 polymorphism and cancer risk in the Caucasian populations (GG vs. AA: OR = 1.72, 95% CI, 1.12-2.64, P = .013, and GG vs. AG/AA: OR = 1.82, 95% CI, 1.19-2.78, P = .006). The potential bias in literature selection was witnessed in this meta-analysis (G vs. A: P Begg's = 0.187, PEgger's = 0.049; and GG/GA vs. AA: P Begg's = 0.069, PEgger's = 0.024). The adjusted ORs and CIs of the nonparametric "trim-and-fill" method demonstrated the reliability of these findings. The outcome of heterogeneity analysis indicated that heterogeneity might be due to small sample sizes (<1000 subjects), cancer types (bladder cancer, other cancers), ethnicity (Asians), and population-based studies. However, the sensitivity analysis validated the reliability of the findings. Conclusion: In conclusion, this updated meta-analysis showed that the G carrier of the MCP-1 gene rs1024611 is associated with susceptibility to cancer in Caucasian.
Collapse
Affiliation(s)
- Zhan Chen
- Department of Cardiothoracic Surgery, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Shiping Yin
- Physical Examination Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Liang Zheng
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Weifeng Tang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Wei Wei
- Department of Cardiothoracic Surgery, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Kang Sui
- Department of Cardiothoracic Surgery, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| |
Collapse
|
31
|
Han Y, Li X, Zhang X, Gao Y, Qi R, Cai R, Qi Y. Isodeoxyelephantopin, a sesquiterpene lactone from Elephantopus scaber Linn., inhibits pro-inflammatory mediators' production through both NF-κB and AP-1 pathways in LPS-activated macrophages. Int Immunopharmacol 2020; 84:106528. [PMID: 32335480 DOI: 10.1016/j.intimp.2020.106528] [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: 03/01/2020] [Revised: 04/11/2020] [Accepted: 04/17/2020] [Indexed: 12/19/2022]
Abstract
Isodeoxyelephantopin (IDET) has been identified as an anti-tumor natural constituent whose anti-tumor activity and mechanism have been widely investigated. Since the occurrence and development of cancer usually accompany with inflammation, and tumor signaling shares many components with inflammation signaling, the agents with anti-tumor activity are likely to possess anti-inflammation potential. Thus, the current study aims to demonstrate the anti-inflammatory activity along with the underlying mechanism of IDET in lipopolysaccharide (LPS)-primed macrophages. By using Griess method and ELISA, we found that in both bone marrow derived macrophages and alveolar macrophage cell line, IDET, at relatively low concentrations (0.75, 1.5 and 3 μM), could inhibit LPS-induced expression of various pro-inflammatory mediators including nitric oxide (NO) generated by inducible nitric oxide synthase (iNOS), interleukin (IL)-6, monocyte chemotactic protein-1 (MCP-1) and IL-1β. Meanwhile, in activated MH-S cells, the inhibitory action of IDET on mRNA expression levels of these cytokines was also detected using qPCR. Mechanistically, the effects of IDET on two key inflammatory signalings, nuclear factor-κB (NF-κB) and activator protein-1 (AP-1) pathways, were determined in LPS-activated MH-S cells by reporter gene along with western blot assays. On the one hand, IDET suppressed NF-κB signaling via down-regulating phosphorylation and degradation of inhibitor of NF-κB (IκB)-α and the subsequent p65 translocation. On the other hand, IDET dampened AP-1 signaling through attenuating phosphorylation of both c-jun N-terminal kinase 1/2 (JNK1/2) and extracellular signal regulated kinase 1/2 (ERK1/2). Our study indicates that IDET might be a promising constituent from the anti-inflammatory herb Elephantopus scaber Linn. in mitigating inflammatory conditions, especially respiratory inflammation.
Collapse
Affiliation(s)
- Yixin Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Ximeng Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Xiaoyu Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yuan Gao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Ruijuan Qi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Runlan Cai
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yun Qi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China.
| |
Collapse
|
32
|
Lima ÉDA, Cavalcante-Silva LHA, Carvalho DCM, Netto CD, Costa PRR, Rodrigues-Mascarenhas S. The pterocarpanquinone LQB 118 inhibits inflammation triggered by zymosan in vivo and in vitro. Int Immunopharmacol 2020; 83:106399. [PMID: 32193104 DOI: 10.1016/j.intimp.2020.106399] [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: 10/15/2019] [Revised: 03/03/2020] [Accepted: 03/10/2020] [Indexed: 11/27/2022]
Abstract
LQB 118, a hydride molecule, has been described as an antineoplastic and antiparasitic drug. Recently, LQB118 was also shown to display anti-inflammatory properties using an LPS-induced lung inflammation model. However, LQB 118 effects on the inflammatory response induced by zymosan has not been demonstrated. In this study, swiss mice were LQB 118 intraperitoneally (i.p.) treated and zymosan was used to induce peritoneal inflammation. Peritoneal fluid was collected and used for cell counting and proinflammatory cytokines quantification (IL-1β, IL-6, and TNF-α) by immunoenzymatic assay (ELISA). For in vitro studies, peritoneal macrophages zymosan-stimulated were used. Results demonstrated that LQB 118 treatment reduced polymorphonuclear cell migration and TNF-α, IL-1β, and IL-6 levels in the peritoneal cavity. In macrophages, LQB 118 treatment display no cytotoxic effect and is also able to reduce cytokines levels. To investigate LQB 118 putative mechanism of action, TLR2, CD69, and P-p38 MAPK expression were evaluated. LQB 118 treatment reduced CD69 expression and p38 phosphorylation induced by zymosan. Furthermore, LQB 118 was able to negatively modulate TLR2 expression in the presence of inflammatory stimulus. Thus, our study provide new evidences for the mechanisms related to the anti-inflammatory effect of LQB 118 in vivo and in vitro.
Collapse
Affiliation(s)
| | | | | | - Chaquip Daher Netto
- Laboratory of Chemistry, Federal University of Rio de Janeiro, 27930-560, Brazil
| | - Paulo Roberto Ribeiro Costa
- Laboratory of Bioorganic Chemistry, Natural Products Research Institute, Federal University of Rio de Janeiro, 21941-590, Brazil
| | | |
Collapse
|
33
|
Spix B, Chao YK, Abrahamian C, Chen CC, Grimm C. TRPML Cation Channels in Inflammation and Immunity. Front Immunol 2020; 11:225. [PMID: 32184778 PMCID: PMC7058977 DOI: 10.3389/fimmu.2020.00225] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/28/2020] [Indexed: 12/24/2022] Open
Abstract
Background: In 1883, Ilya Mechnikov discovered phagocytes and established the concept of phagocytosis by macrophages. In 1908, he was awarded the Nobel Prize in Physiology/Medicine for his findings, which laid the foundations for today's understanding of the innate immune response. Only in the 1960s, Max Cooper and Robert Good significantly advanced our understanding of the immune system by demonstrating that B- and T-cells cooperate to regulate the adaptive immune response. Both, innate and adaptive immune response are essential to effectively protect the individual against infectious agents, such as viruses, bacterial or insect toxins, or allergens. Innate immune responses occur rapidly upon exposure to noxious or infectious agents or organisms, in contrast to the adaptive immune system that needs days rather than hours to develop and acts primarily on the basis of antigen-specific receptors expressed on the surface of B- and T-lymphocytes. In recent years, it has become evident that endosomes and lysosomes are involved in many aspects of immune cell function, such as phagocytosis, antigen presentation and processing by antigen-presenting cells, release of proinflammatory mediators, e.g., by mast cells, or secretion of the pore-forming protein perforin by cytotoxic T lymphocytes. Several lysosomal storage disorders (LSDs) have been associated with defects in immune system function or immune system hyperactivity, such as Gaucher, Fabry, or Niemann-Pick type C1 disease, mucopolysaccharidoses (MPS), gangliosidosis, or juvenile neuronal ceroid lipofuscinosis (JNCL). Beside accumulating evidence on the importance of endolysosomes in immune cell function, recent results suggest direct roles of endolysosomal ion channels, such as the TRPML channels (mucolipins), which are members of the transient receptor potential (TRP) superfamily of non-selective cation channels, for different aspects of immune cell function. The aim of this review is to discuss the current knowledge about the roles of TRPML channels in inflammation and immunity, and to assess their potential as drug targets to influence immune cell functions. Advances: Examples of recently established roles of TRPML channels in immune system function and immune response include the TRPML1-mediated modulation of secretory lysosomes, granzyme B content, and tuning of effector function in NK cells, TRPML1-dependent directional dendritic cell (DC) migration and DC chemotaxis, and the role of TRPML2 in chemokine release from LPS-stimulated macrophages. Outlook: Although our understanding of the functional roles of TRPML channels in inflammation and immunity is still in its infancy, a few interesting findings have been made in the past years, encouraging further and more detailed work on the role of TRPMLs, e.g., in intracellular trafficking and release of chemokines, cytokines, or granzyme B, or in phagocytosis and bacterial toxin and virus trafficking through the endolysosomal machinery.
Collapse
Affiliation(s)
- Barbara Spix
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Yu-Kai Chao
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Carla Abrahamian
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Cheng-Chang Chen
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Christian Grimm
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
34
|
Gschwandtner M, Derler R, Midwood KS. More Than Just Attractive: How CCL2 Influences Myeloid Cell Behavior Beyond Chemotaxis. Front Immunol 2019; 10:2759. [PMID: 31921102 PMCID: PMC6923224 DOI: 10.3389/fimmu.2019.02759] [Citation(s) in RCA: 434] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1/CCL2) is renowned for its ability to drive the chemotaxis of myeloid and lymphoid cells. It orchestrates the migration of these cell types both during physiological immune defense and in pathological circumstances, such as autoimmune diseases including rheumatoid arthritis and multiple sclerosis, inflammatory diseases including atherosclerosis, as well as infectious diseases, obesity, diabetes, and various types of cancer. However, new data suggest that the scope of CCL2's functions may extend beyond its original characterization as a chemoattractant. Emerging evidence shows that it can impact leukocyte behavior, influencing adhesion, polarization, effector molecule secretion, autophagy, killing, and survival. The direction of these CCL2-induced responses is context dependent and, in some cases, synergistic with other inflammatory stimuli. The involvement of CCL2 signaling in multiple diseases renders it an interesting therapeutic target, although current targeting strategies have not met early expectations in the clinic. A better understanding of how CCL2 affects immune cells will be pivotal to the improvement of existing therapeutic approaches and the development of new drugs. Here, we provide an overview of the pleiotropic effects of CCL2 signaling on cells of the myeloid lineage, beyond chemotaxis, and highlight how these actions might help to shape immune cell behavior and tumor immunity.
Collapse
Affiliation(s)
- Martha Gschwandtner
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Rupert Derler
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Kim S. Midwood
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
35
|
Elsaafien K, Korim WS, Setiadi A, May CN, Yao ST. Chemoattraction and Recruitment of Activated Immune Cells, Central Autonomic Control, and Blood Pressure Regulation. Front Physiol 2019; 10:984. [PMID: 31427987 PMCID: PMC6688384 DOI: 10.3389/fphys.2019.00984] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/15/2019] [Indexed: 12/16/2022] Open
Abstract
Inflammatory mediators play a critical role in the regulation of sympathetic outflow to cardiovascular organs in hypertension. Emerging evidence highlights the involvement of immune cells in the regulation of blood pressure. However, it is still unclear how these immune cells are activated and recruited to key autonomic brain regions to regulate sympathetic outflow to cardiovascular organs. Chemokines such as C-C motif chemokine ligand 2 (CCL2), and pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1β), are upregulated both peripherally and centrally in hypertension. More specifically, they are upregulated in key autonomic brain regions that control sympathetic activity and blood pressure such as the paraventricular nucleus of the hypothalamus and the rostral ventrolateral medulla. Furthermore, this upregulation of inflammatory mediators is associated with the infiltration of immune cells to these brain areas. Thus, expression of pro-inflammatory chemokines and cytokines is a potential mechanism promoting invasion of immune cells into key autonomic brain regions. In pathophysiological conditions, this can result in abnormal activation of brain circuits that control sympathetic nerve activity to cardiovascular organs and ultimately in increases in blood pressure. In this review, we discuss emerging evidence that helps explain how immune cells are chemoattracted to autonomic nuclei and contribute to changes in sympathetic outflow and blood pressure.
Collapse
Affiliation(s)
- Khalid Elsaafien
- Discovery Science, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Willian S. Korim
- Discovery Science, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Anthony Setiadi
- Discovery Science, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Clive N. May
- Discovery Science, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Song T. Yao
- Discovery Science, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
36
|
Marinobufagenin Inhibits Neutrophil Migration and Proinflammatory Cytokines. J Immunol Res 2019; 2019:1094520. [PMID: 31236418 PMCID: PMC6545758 DOI: 10.1155/2019/1094520] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/28/2019] [Indexed: 12/12/2022] Open
Abstract
Cardiotonic steroids, such as ouabain and digoxin, are known to bind to Na+/K+-ATPase and to promote several biological activities, including anti-inflammatory activity. However, there are still no reports in the literature about inflammation and marinobufagenin, a cardiotonic steroid from the bufadienolide family endogenously found in mammals. Therefore, the aim of this work was to analyze, in vivo and in vitro, the role of marinobufagenin in acute inflammation. Swiss mice were treated with 0.56 mg/kg of marinobufagenin intraperitoneally (i.p.) and zymosan (2 mg/mL, i.p.) was used to induce peritoneal inflammation. Peritoneal fluid was collected and used for counting cells by optical microscopy and proinflammatory cytokine quantification (IL-1β, IL-6, and TNF-α) by immunoenzymatic assay (ELISA). Zymosan stimulation, as expected, induced increased cell migration and proinflammatory cytokine levels in the peritoneum. Marinobufagenin treatment reduced polymorphonuclear cell migration and IL-1β and IL-6 levels in the peritoneal cavity, without interfering in TNF-α levels. In addition, the effect of marinobufagenin was evaluated using peritoneal macrophages stimulated by zymosan (0.2 mg/mL) in vitro. Marinobufagenin treatment at different concentrations (10, 100, 1000, and 10000 nM) showed no cytotoxic effect on peritoneal macrophages. Interestingly, the lowest concentration, which did not inhibit Na+/K+-ATPase activity, attenuated proinflammatory cytokines IL-1β, IL-6, and TNF-α levels. To investigate the putative mechanism of action of marinobufagenin, the expression of surface molecules (TLR2 and CD69) and P-p38 MAPK were also evaluated, but no significant effect was observed. Thus, our results suggest that marinobufagenin has an anti-inflammatory role in vivo and in vitro and reveals a novel possible endogenous function of this steroid in mammals.
Collapse
|
37
|
Luissint AC, Williams HC, Kim W, Flemming S, Azcutia V, Hilgarth RS, Leary MNO, Denning TL, Nusrat A, Parkos CA. Macrophage-dependent neutrophil recruitment is impaired under conditions of increased intestinal permeability in JAM-A-deficient mice. Mucosal Immunol 2019; 12:668-678. [PMID: 30745566 PMCID: PMC6543824 DOI: 10.1038/s41385-019-0143-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023]
Abstract
Junctional adhesion molecule-A (JAM-A) is a transmembrane glycoprotein expressed on leukocytes, endothelia, and epithelia that regulates biological processes including barrier function and immune responses. While JAM-A has been reported to facilitate tissue infiltration of leukocytes under inflammatory conditions, the contributions of leukocyte-expressed JAM-A in vivo remain unresolved. We investigated the role of leukocyte-expressed JAM-A in acute peritonitis induced by zymosan, lipopolysaccharide (LPS), or TNFα using mice with selective loss of JAM-A in myelomonocytic cells (LysM-Cre;Jam-afl/fl). Surprisingly, in LysM-Cre;Jam-afl/fl mice, loss of JAM-A did not affect neutrophil (PMN) recruitment into the peritoneum in response to zymosan, LPS, or TNFα although it was significantly reduced in Jam-aKO mice. In parallel, Jam-aKO peritoneal macrophages exhibited diminished CXCL1 chemokine production and decreased activation of NF-kB, whereas those from LysM-Cre;Jam-afl/fl mice were unaffected. Using Villin-Cre;Jam-afl/fl mice, targeted loss of JAM-A on intestinal epithelial cells resulted in increased intestinal permeability along with reduced peritoneal PMN migration as well as lower levels of CXCL1 and active NF-kB similar to that observed in Jam-aKO animals. Interestingly, in germ-free Villin-Cre;Jam-afl/fl mice, PMN recruitment was unaffected suggesting dependence on gut microbiota. Such observations highlight the functional link between a leaky gut and regulation of innate immune responses.
Collapse
Affiliation(s)
| | - Holly C Williams
- Department of Medicine, Division of Cardiology, Emory University, Atlanta, GA, USA
| | - Wooki Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin, Korea
| | - Sven Flemming
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Veronica Azcutia
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Timothy L Denning
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
38
|
Abstract
The distribution of dust particles within the lungs and their excretion are highly associated with their pulmonary toxicity. Literature was reviewed to discern pulmonary translocation pathways for inhaled α-quartz compared to those for inhaled TiO2. Accordingly, it was hypothesized α-quartz particles in the alveoli were phagocytized by alveolar macrophages but silica-containing macrophages remained in the alveoli for longer time in contrast to the rapid elimination from the alveoli seen for TiO2-containing macrophages. In addition, it was presumed that free silica particles are translocated in the interstitium, possibly through the cytoplasm of Type I epithelial cells, as observed with TiO2. Free silica particles are presumed to be phagocytized by interstitial macrophages soon after the particles penetrate the interstitium; these dust cells are then translocated to the ciliated airway regions in the lumen through bronchus-associated lymphoid tissue (BALT). The pulmonary retention half-time of dust particles in rats exposed to α-quartz is several times longer than that of rats exposed to TiO2, as long as the lung dust burden is ≈ 3 mg. The reduced pulmonary particle clearance ability in rats exposed to α-quartz aerosol is presumably attributed to the long-term retention of dust cells both in the alveoli and in the interstitium; this retention may be caused by the reduced chemotactic abilities of α-quartz-containing dust cells. However, the accumulation of α-quartz-containing dust cells in the lungs is not associated with the occurrence of pulmonary inflammation.
Collapse
|
39
|
Zhang K, Luo J. Role of MCP-1 and CCR2 in alcohol neurotoxicity. Pharmacol Res 2019; 139:360-366. [PMID: 30472461 PMCID: PMC6360095 DOI: 10.1016/j.phrs.2018.11.030] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/22/2018] [Accepted: 11/21/2018] [Indexed: 01/08/2023]
Abstract
Alcohol abuse causes profound damage to both the developing brain and the adult brain. Prenatal exposure to alcohol results in a wide range of deficits known as fetal alcohol spectrum disorders (FASD). Alcohol abuse in adults is associated with brain shrinkage, memory and attention deficits, communication disorders and physical disabilities. Monocyte chemoattractant protein-1 (MCP-1/CCL2) is one of the key chemokines that regulate the recruitment and activation of monocytes and microglia. Both MCP-1 and its receptor C-C chemokine receptor type 2 (CCR2) expressed in the brain are involved in various neuroinflammatory disorders, such as multiple sclerosis (MS), Alzheimer's disease (AD) and Parkinson's disease (PD). However, the role of MCP-1/CCR2 in alcohol-induced brain damage is unclear. Recent evidence indicates that alcohol exposure increased the activity of MCP-1/CCR2 in both mature and developing central nervous systems (CNS). MCP-1/CCR2 signaling in the brain was involved in alcohol drinking behavior. MCP-1/CCR2 inhibition alleviated alcohol neurotoxicity by reducing microglia activation/neuroinflammation in the developing brain and spinal cord. In this review, we discussed the role of MCP-1/CCR2 signaling in alcohol-induced neuroinflammation and brain damage. We also discussed the signaling cascades that are involved in the activation of MCP-1/CCR2 in response to alcohol exposure.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Lexington VA Health Care System, Research & Development, 1101 Veterans Drive, Lexington, KY 40502, USA.
| |
Collapse
|
40
|
DDX3 Participates in Translational Control of Inflammation Induced by Infections and Injuries. Mol Cell Biol 2018; 39:MCB.00285-18. [PMID: 30373933 DOI: 10.1128/mcb.00285-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/10/2018] [Indexed: 12/28/2022] Open
Abstract
Recent studies have suggested that DDX3 functions in antiviral innate immunity, but the underlying mechanism remains elusive. We previously identified target mRNAs whose translation is controlled by DDX3. Pathway enrichment analysis of these targets indicated that DDX3 is involved in various infections and inflammation. Using immunoblotting, we confirmed that PACT, STAT1, GNB2, Rac1, TAK1, and p38 mitogen-activated protein kinase (MAPK) proteins are downregulated by DDX3 knockdown in human monocytic THP-1 cells and epithelial HeLa cells. Polysome profiling revealed that DDX3 knockdown reduces the translational efficiency of target mRNAs. We further demonstrated DDX3-mediated translational control of target mRNAs by luciferase reporter assays. To examine the effects of DDX3 knockdown on macrophage migration and phagocytosis, we performed in vitro cell migration assay and flow cytometry analysis of the uptake of green fluorescent protein-expressing Escherichia coli in THP-1 cells. The DDX3 knockdown cells exhibited impaired macrophage migration and phagocytosis. Moreover, we used a human cytokine antibody array to identify the cytokines affected by DDX3 knockdown. Several chemokines were decreased considerably in DDX3 knockdown THP-1 cells after lipopolysaccharide or poly(I·C) stimulation. Lastly, we demonstrated that DDX3 is crucial for the recruitment of phagocytes to the site of inflammation in transgenic zebrafish.
Collapse
|
41
|
Schwager S, Renner S, Hemmerle T, Karaman S, Proulx ST, Fetz R, Golding-Ochsenbein AM, Probst P, Halin C, Neri D, Detmar M. Antibody-mediated delivery of VEGF-C potently reduces chronic skin inflammation. JCI Insight 2018; 3:124850. [PMID: 30518687 DOI: 10.1172/jci.insight.124850] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/24/2018] [Indexed: 02/06/2023] Open
Abstract
VEGF-C is an important mediator of lymphangiogenesis and has been shown to alleviate chronic inflammation in a variety of disease models. In this study, we investigated whether targeted delivery of VEGF-C to sites of inflammation and site-specific activation of lymphatic vessels would represent a clinically feasible strategy for treating chronic skin inflammation. To this end, we generated a fusion protein consisting of human VEGF-C fused to the F8 antibody (F8-VEGF-C), which is specific for the alternatively spliced, angiogenesis-marking extradomain A (EDA) of fibronectin. In two mouse models of psoriasis-like skin inflammation, mediated by transgenic VEGF-A overexpression or repeated application of imiquimod, intravenous treatment with F8-VEGF-C but not with untargeted VEGF-C significantly reduced ear skin edema and was as effective as the clinically used TNF-α receptor-Fc fusion protein (TNFR-Fc). Treatment with F8-VEGF-C led to a marked expansion of lymphatic vessels in the inflamed skin and significantly improved lymphatic drainage function. At the same time, treatment with F8-VEGF-C significantly reduced leukocyte numbers, including CD4+ and γδ T cells. In sum, our results reveal that targeted delivery of VEGF-C and site-specific induction of lymphatic vessels represent a potentially new and promising approach for the treatment of chronic inflammatory diseases.
Collapse
|
42
|
Molecular Imaging of a New Multimodal Microbubble for Adhesion Molecule Targeting. Cell Mol Bioeng 2018; 12:15-32. [PMID: 31719897 PMCID: PMC6816780 DOI: 10.1007/s12195-018-00562-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 11/09/2018] [Indexed: 12/29/2022] Open
Abstract
Introduction Inflammation is an important risk-associated component of many diseases and can be diagnosed by molecular imaging of specific molecules. The aim of this study was to evaluate the possibility of targeting adhesion molecules on inflammation-activated endothelial cells and macrophages using an innovative multimodal polyvinyl alcohol-based microbubble (MB) contrast agent developed for diagnostic use in ultrasound, magnetic resonance, and nuclear imaging. Methods We assessed the binding efficiency of antibody-conjugated multimodal contrast to inflamed murine or human endothelial cells (ECs), and to peritoneal macrophages isolated from rats with peritonitis, utilizing the fluorescence characteristics of the MBs. Single-photon emission tomography (SPECT) was used to illustrate 99mTc-labeled MB targeting and distribution in an experimental in vivo model of inflammation. Results Flow cytometry and confocal microscopy showed that binding of antibody-targeted MBs to the adhesion molecules ICAM-1, VCAM-1, or E-selectin, expressed on cytokine-stimulated ECs, was up to sixfold higher for human and 12-fold higher for mouse ECs, compared with that of non-targeted MBs. Under flow conditions, both VCAM-1- and E-selectin-targeted MBs adhered more firmly to stimulated human ECs than to untreated cells, while VCAM-1-targeted MBs adhered best to stimulated murine ECs. SPECT imaging showed an approximate doubling of signal intensity from the abdomen of rats with peritonitis, compared with healthy controls, after injection of anti-ICAM-1-MBs. Conclusions This novel multilayer contrast agent can specifically target adhesion molecules expressed as a result of inflammatory stimuli in vitro, and has potential for use in disease-specific multimodal diagnostics in vivo using antibodies against targets of interest.
Collapse
|
43
|
|
44
|
Zika plasma viral dynamics in nonhuman primates provides insights into early infection and antiviral strategies. Proc Natl Acad Sci U S A 2017; 114:8847-8852. [PMID: 28765371 DOI: 10.1073/pnas.1704011114] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The recent outbreak of Zika virus (ZIKV) has been associated with fetal abnormalities and neurological complications, prompting global concern. Here we present a mathematical analysis of the within-host dynamics of plasma ZIKV burden in a nonhuman primate model, allowing for characterization of the growth and clearance of ZIKV within individual macaques. We estimate that the eclipse phase for ZIKV, the time between cell infection and viral production, is most likely short (∼4 h), the median within-host basic reproductive number R0 is 10.7, the rate of viral production is rapid (>25,000 virions d-1), and the lifetime of an infected cell while producing virus is ∼5 h. We also estimate that the minimum number of virions produced by an infected cell over its lifetime is ∼5,500. We assess the potential effect of an antiviral treatment that blocks viral replication, showing that the median time to undetectable plasma viral load (VL) can be reduced from ∼5 d to ∼3 d with a drug concentration ∼15 times the drug's EC50 when treatment is given prophylactically starting at the time of infection. In the case of favipiravir, a polymerase inhibitor with activity against ZIKV, we predict a dose of 150 mg/kg given twice a day initiated at the time of infection can reduce the peak median VL by ∼3 logs and shorten the time to undetectable median VL by ∼2 d, whereas treatment given 2 d postinfection is mostly ineffective in accelerating plasma VL loss in macaques.
Collapse
|
45
|
Lin HC, Ho MY, Tsen CM, Huang CC, Wu CC, Huang YJ, Hsiao IL, Chuang CY. From the Cover: Comparative Proteomics Reveals Silver Nanoparticles Alter Fatty Acid Metabolism and Amyloid Beta Clearance for Neuronal Apoptosis in a Triple Cell Coculture Model of the Blood–Brain Barrier. Toxicol Sci 2017; 158:151-163. [DOI: 10.1093/toxsci/kfx079] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
46
|
Nandi A, Bishayi B. CCR-2 neutralization augments murine fresh BMC activation by Staphylococcus aureus via two distinct mechanisms: at the level of ROS production and cytokine response. Innate Immun 2017; 23:345-372. [PMID: 28409543 DOI: 10.1177/1753425917697806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
CCR-2 signaling regulates recruitment of monocytes from the bone marrow into the bloodstream and then to sites of infection. We sought to determine whether CCL-2/CCR-2 signaling is involved in the killing of Staphylococcus aureus by murine bone marrow cells (BMCs). The intermittent link of reactive oxygen species (ROS)-NF-κB/p38-MAPK-mediated CCL-2 production in CCR-2 signaling prompted us to determine whether neutralization of CCR-2 augments the response of murine fresh BMCs (FBMCs) after S. aureus infection. It was observed that anti-CCR-2 Ab-treated FBMCs released fewer ROS on encountering S. aureus infection than CCR-2 non-neutralized FBMCs, also correlating with reduced killing of S. aureus in CCR-2 neutralized FBMCs. Staphylococcal catalase and SOD were also found to play a role in protecting S. aureus from the ROS-mediated killing of FBMC. S. aureus infection of CCR-2 intact FBMCs pre-treated with either NF-κB or p-38-MAPK blocker induced less CCL-2, suggesting that NF-κB or p-38-MAPK is required for CCL-2 production by FBMCs. Moreover, blocking of CCR-2 along with NF-κB or p-38-MAPK resulted in elevated CCL-2 production and reduced CCR-2 expression. Inhibition of CCR-2 impairs the response of murine BMCs to S. aureus infection by attenuation ROS production and modulating the cytokine response.
Collapse
Affiliation(s)
- Ajeya Nandi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, West Bengal, India
| |
Collapse
|
47
|
Raza A, Firasat S, Khaliq S, Khan AR, Mahmood S, Aziz T, Mubarak M, Naqvi SAA, Rizvi SAH, Abid A. Monocyte Chemoattractant Protein-1 (MCP-1/CCL2) Levels and Its Association with Renal Allograft Rejection. Immunol Invest 2017; 46:251-262. [PMID: 27960564 DOI: 10.1080/08820139.2016.1248559] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND CCL2 is a chemoattractant for monocytes/macrophages, T cells, and natural killer cells. It is shown to be involved in the immunological responses against renal allograft. This study was conducted to access the role of urinary CCL2 expression in predicting the rejection episodes in renal transplant patients. METHOD A total of 409 urine samples included in this study. The samples were consisted of (a) biopsy-proven graft rejection (n = 165); (b) non-rejection (n = 93); (c) non-biopsy stable-graft (n = 42), and (d) healthy renal donors (n = 109). The samples were quantified for the CCL2 using the MCP-1/CCL2 ELISA kit. The data were analyzed using the Statistical Package for Social Sciences (SPSS®) and MedCalc® statistical software. RESULTS Results showed that the CCL2 levels were significantly increased in rejection group when compared with the non-rejection, stable-graft, and control, P < 0.05. The receiver operating curve's characteristics illustrated that the urinary CCL2 level is a good predictor for graft rejection, with an area under the curve of 0.81 ± 0.03 with optimum sensitivity and specificity of 87% and 62%, respectively, at a cut-off value of 198 pg/mL. Kaplan-Meier curve also showed better cumulative rejection-free graft survival time in group with less than 198 pg/mL of CCL2 as compared to those with expression levels of more than 198 pg/mL (30 weeks vs. 3 weeks; log-rank test, P < 0.001). CONCLUSION In our study, noninvasive investigation of CCL2 levels in urine has showed potential to predict rejection episodes. It is suggested that the CCL2, with others markers, may help in early detection and monitoring of graft rejection episodes.
Collapse
Affiliation(s)
- Ali Raza
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Sadaf Firasat
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Shagufta Khaliq
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
- b Department of Human Genetics , University of Health Sciences , Lahore , Pakistan
| | - Abdul Rafay Khan
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Shafaq Mahmood
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Tahir Aziz
- c Department of Urology , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Muhammad Mubarak
- d Department of Histopathology , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Syed Ali Anwar Naqvi
- c Department of Urology , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Syed Adibul Hasan Rizvi
- c Department of Urology , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| | - Aiysha Abid
- a Centre for Human Genetics and Molecular Medicine , Sindh Institute of Urology and Transplantation , Karachi , Pakistan
| |
Collapse
|
48
|
Rezar-Dreindl S, Eibenberger K, Pollreisz A, Bühl W, Georgopoulos M, Krall C, Dunavölgyi R, Weigert G, Kroh ME, Schmidt-Erfurth U, Sacu S. Effect of intravitreal dexamethasone implant on intra-ocular cytokines and chemokines in eyes with retinal vein occlusion. Acta Ophthalmol 2017; 95:e119-e127. [PMID: 27417275 DOI: 10.1111/aos.13152] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/04/2016] [Indexed: 12/27/2022]
Abstract
PURPOSE To investigate the influence of intravitreal dexamethasone implant on inflammatory and angiogenic cytokine levels in the aqueous of patients with retinal vein occlusion (RVO). METHODS Forty eyes of 40 consecutive patients with macular oedema (ME) due to branch and central retinal vein occlusion (BRVO/CRVO) were treated with an intravitreal dexamethasone implant (Ozurdex® ) at baseline and evaluated until month 6. Retreatment was performed in case of recurrent ME earliest 4 months after the baseline treatment. Aqueous humour samples were taken at baseline, months 1, 3, 6 and at the time of each retreatment. Concentrations of 29 different cytokines were measured by Luminex® bead assays. The control group comprised healthy patients undergoing cataract surgery. RESULTS At baseline concentrations of interleukin (IL)-8, angiopoietin (ANG)-2 and intercellular adhesion molecule (ICAM)-1 were highly elevated in patients with CRVO compared with controls (p = 0.006; p = 0.02; p = 0.03). Vascular endothelial growth factor (VEGF) concentrations were upregulated in patients with BRVO and CRVO (p = 0.003; p = 0.001). Retreatment with a dexamethasone implant was necessary after 4 months in 14/8 (BRVO/CRVO) patients, 5 months in 5/3 patients and 6 months in one patient (BRVO). After the initial treatment, macrophage chemo-attractant protein (MCP)-1 and IL17-E concentrations decreased in BRVO (p < 0.001; p = 0.01) and MCP-1 and IL1-α in CRVO (p = 0.01; p = 0.003). Vascular endothelial growth factor (VEGF) concentrations did not change during treatment in either group (p = 0.3). A mixed-effect model showed that cytokine concentrations positively correlated with central retinal thickness changes. CONCLUSIONS Intravitreal dexamethasone treatment resulted in alterations in the concentrations of pro-inflammatory cytokines MCP-1 and IL17-E in patients with BRVO and MCP-1 and IL1-α in patients with CRVO. These data highlight the important role of inflammatory mediators involved in ME due to RVO.
Collapse
Affiliation(s)
| | | | - Andreas Pollreisz
- Department of Ophthalmology; Medical University of Vienna; Vienna VIE Austria
| | - Wolf Bühl
- Department of Ophthalmology; Medical University of Vienna; Vienna VIE Austria
| | | | - Christoph Krall
- Department of Medical Statistics; Medical University of Vienna; Vienna VIE Austria
| | - Roman Dunavölgyi
- Department of Ophthalmology; Medical University of Vienna; Vienna VIE Austria
| | - Günther Weigert
- Department of Ophthalmology; Medical University of Vienna; Vienna VIE Austria
| | | | | | - Stefan Sacu
- Department of Ophthalmology; Medical University of Vienna; Vienna VIE Austria
| |
Collapse
|
49
|
Groeger SE, Meyle J. Epithelial barrier and oral bacterial infection. Periodontol 2000 2017; 69:46-67. [PMID: 26252401 DOI: 10.1111/prd.12094] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2015] [Indexed: 01/11/2023]
Abstract
The oral epithelial barrier separates the host from the environment and provides the first line of defense against pathogens, exogenous substances and mechanical stress. It consists of underlying connective tissue and a stratified keratinized epithelium with a basement membrane, whose cells undergo terminal differentiation resulting in the formation of a mechanically resistant surface. Gingival keratinocytes are connected by various transmembrane proteins, such as tight junctions, adherens junctions and gap junctions, each of which has a specialized structure and specific functions. Periodontal pathogens are able to induce inflammatory responses that lead to attachment loss and periodontal destruction. A number of studies have demonstrated that the characteristics of pathogenic oral bacteria influence the expression and structural integrity of different cell-cell junctions. Tissue destruction can be mediated by host cells following stimulation with cytokines and bacterial products. Keratinocytes, the main cell type in gingival epithelial tissues, express a variety of proinflammatory cytokines and chemokines, including interleukin-1alpha, interleukin-1beta, interleukin-6, interleukin-8 and tumor necrosis factor-alpha. Furthermore, the inflammatory mediators that may be secreted by oral keratinocytes are vascular endothelial growth factor, prostaglandin E2 , interleukin-1 receptor antagonist and chemokine (C-C motif) ligand 2. The protein family of matrix metalloproteinases is able to degrade all types of extracellular matrix protein, and can process a number of bioactive molecules. Matrix metalloproteinase activities under inflammatory conditions are mostly deregulated and often increased, and those mainly relevant in periodontal disease are matrix metalloproteinases 1, 2, 3, 8, 9, 13 and 24. Viral infection may also influence the epithelial barrier. Studies show that the expression of HIV proteins in the mucosal epithelium is correlated with the disruption of epithelial tight junctions, suggesting a possible enhancement of human papilloma virus infection by HIV-associated disruption of tight junctions. Altered expression of matrix metalloproteinases was demonstrated in keratinocytes transformed with human papilloma virus-16 or papilloma virus-18,. To summarize, the oral epithelium is able to react to a variety of exogenous, possibly noxious influences.
Collapse
|
50
|
Siddiqui JA, Partridge NC. CCL2/Monocyte Chemoattractant Protein 1 and Parathyroid Hormone Action on Bone. Front Endocrinol (Lausanne) 2017; 8:49. [PMID: 28424660 PMCID: PMC5372820 DOI: 10.3389/fendo.2017.00049] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/27/2017] [Indexed: 11/13/2022] Open
Abstract
Chemokines are small molecules that play a crucial role as chemoattractants for several cell types, and their components are associated with host immune responses and repair mechanisms. Chemokines selectively recruit monocytes, neutrophils, and lymphocytes and induce chemotaxis through the activation of G protein-coupled receptors. Two well-described chemokine families (CXC and CC) are known to regulate the localization and trafficking of immune cells in cases of injury, infection, and tumors. Monocyte chemoattractant protein 1 (MCP-1/CCL2) is one of the important chemokines from the CC family that controls migration and infiltration of monocytes/macrophages during inflammation. CCL2 is profoundly expressed in osteoporotic bone and prostate cancer-induced bone resorption. CCL2 also regulates physiological bone remodeling in response to hormonal and mechanical stimuli. Parathyroid hormone (PTH) has multifaceted effects on bone, depending on the mode of administration. Intermittent PTH increases bone in vivo by increasing the number and activity of osteoblasts, whereas a continuous infusion of PTH decreases bone mass by stimulating a net increase in bone resorption. CCL2 is essential for both anabolic and catabolic effects of PTH. In this review, we will discuss the pharmacological role of PTH and involvement of CCL2 in the processes of PTH-mediated bone remodeling.
Collapse
Affiliation(s)
- Jawed Akhtar Siddiqui
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - Nicola C. Partridge
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
- *Correspondence: Nicola C. Partridge,
| |
Collapse
|