1
|
AbdelRazic MI, Hakeem GLA, Hanna MS, Mohamed OM, Abuelela IS. Enhanced mortality prediction in pediatric sepsis using NGAL: A comparison with PRISM III scores in critical care settings. Eur J Pediatr 2025; 184:201. [PMID: 39954103 PMCID: PMC11829929 DOI: 10.1007/s00431-025-06017-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/17/2025] [Accepted: 01/29/2025] [Indexed: 02/17/2025]
Abstract
Sepsis is a critical condition that disrupts metabolic, physiological, and immune functions, often resulting in severe complications such as multi-organ failure and increased mortality. Neutrophil gelatinase-associated lipocalin (NGAL) has emerged as a promising biomarker for infection and inflammation, offering potential advantages for early mortality prediction. This study compared the predictive value of serum NGAL levels with pediatric risk of mortality III (PRISM III) scores in critically ill pediatric patients with sepsis. A prospective cohort study was conducted at a tertiary hospital from September 2022 to March 2023, involving 75 pediatric patients diagnosed with sepsis, septic shock, or multi-organ dysfunction syndrome (MODS), along with 25 healthy controls. Serum NGAL levels were measured within the first hour of PICU admission and analyzed alongside PRISM III scores to evaluate their correlation with mortality and sepsis severity. The results demonstrated that serum NGAL levels were significantly elevated in septic patients compared to controls, with the highest levels observed in those with MODS. NGAL showed greater sensitivity and specificity for predicting mortality than PRISM III scores, with ROC curve analysis revealing that NGAL levels > 599 mg/ml were strongly associated with increased mortality risk (sensitivity 70.4% and specificity 50%). Multivariate analysis confirmed NGAL as an independent predictor of mortality, outperforming PRISM III scores in identifying severe cases. CONCLUSION Serum NGAL is a valuable biomarker for early prediction of mortality and sepsis severity in pediatric patients, providing faster and more accurate assessments than PRISM III scores. Its integration into clinical practice may enhance decision-making in pediatric critical care settings, allowing for timely interventions and improved patient outcomes. WHAT IS KNOWN • Pediatric risk of mortality III (Prism III) scores is widely used to predict sepsis severity and mortality in pediatric intensive care units, but requiring 12-24 hours to complete. Neutrophil is an established biomarker for inflammation and infection with a potentially anti-pathological value in the neutrophil gelatinus-lipocalin (NGAL) sepsis. WHAT IS NEW • Serum NGAL levels, PICU is measured within the first hour of entry, prism III score in pediatric patients in predicting mortality and severity of sepsis. > An NGAL cutoff of 599 mg/mL is significantly associated with mortality risk, which provides a rapid, independent and more immediate immunity tool for important care decision making.
Collapse
Affiliation(s)
| | | | - Mina Sobhy Hanna
- Pediatrics Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - Omima M Mohamed
- Clinical Pathology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | | |
Collapse
|
2
|
Che R, Wang Q, Li M, Shen J, Ji J. Quantitative Proteomics of Tissue-Infiltrating T Cells From CRC Patients Identified Lipocalin-2 Induces T-Cell Apoptosis and Promotes Tumor Cell Proliferation by Iron Efflux. Mol Cell Proteomics 2024; 23:100691. [PMID: 38072118 PMCID: PMC10792491 DOI: 10.1016/j.mcpro.2023.100691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/25/2023] [Accepted: 12/07/2023] [Indexed: 01/02/2024] Open
Abstract
T cells play the most pivotal roles in antitumor immunity; the T-cell proteome and the differentially expressed proteins in the tumor immune microenvironment have rarely been identified directly from the clinical samples, especially for tumors that lack effective immunotherapy targets, such as colorectal cancer (CRC). In this study, we analyzed the protein expression pattern of the infiltrating T cells isolated from CRC patients using quantitative proteomics. CD4+ and CD8+ T cells were isolated from clinical samples and labeled by tandem mass tag reagents, and the differentially expressed proteins were quantified by mass spectrometry. The T-cell proteome profiling revealed dysfunctions in these tumor-infiltrating T cells. Specifically, antitumor immunity was suppressed because of differentially expressed metal ion transporters and immunity regulators. For the first time, lipocalin-2 (LCN2) was shown to be significantly upregulated in CD4+ T cells. Quantitative proteomic analysis of LCN2-overexpressed Jurkat cells showed that LCN2 damaged T cells by changes in iron transport. LCN2 induced T-cell apoptosis by reducing cellular iron concentration; moreover, the iron that was transported to the tumor microenvironment aided tumor cell proliferation, promoting tumor development. Meanwhile, LCN2 also influenced tumor progression through immune cytokines and cholesterol metabolism. Our results demonstrated that LCN2 has immunosuppressive functions that can promote tumor development; therefore, it is a potential immunotherapy target for CRC.
Collapse
Affiliation(s)
- Rui Che
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Qingsong Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Minzhe Li
- General Surgery Department, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jian Shen
- General Surgery Department, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Jianguo Ji
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
3
|
Romejko K, Markowska M, Niemczyk S. The Review of Current Knowledge on Neutrophil Gelatinase-Associated Lipocalin (NGAL). Int J Mol Sci 2023; 24:10470. [PMID: 37445650 DOI: 10.3390/ijms241310470] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) is a 25-kDa protein that is secreted mostly by immune cells such as neutrophils, macrophages, and dendritic cells. Its production is stimulated in response to inflammation. The concentrations of NGAL can be measured in plasma, urine, and biological fluids such as peritoneal effluent. NGAL is known mainly as a biomarker of acute kidney injury and is released after tubular damage and during renal regeneration processes. NGAL is also elevated in chronic kidney disease and dialysis patients. It may play a role as a predictor of the progression of renal function decreases with complications and mortality due to kidney failure. NGAL is also useful in the diagnostic processes of cardiovascular diseases. It is highly expressed in injured heart tissue and atherosclerostic plaque; its serum concentrations correlate with the severity of heart failure and coronary artery disease. NGAL increases inflammatory states and its levels rise in arterial hypertension, obesity, diabetes, and metabolic complications such as insulin resistance, and is also involved in carcinogenesis. In this review, we present the current knowledge on NGAL and its involvement in different pathologies, especially its role in renal and cardiovascular diseases.
Collapse
Affiliation(s)
- Katarzyna Romejko
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine-National Research Institute, 128 Szaserów Street, 04-141 Warsaw, Poland
| | - Magdalena Markowska
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine-National Research Institute, 128 Szaserów Street, 04-141 Warsaw, Poland
| | - Stanisław Niemczyk
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine-National Research Institute, 128 Szaserów Street, 04-141 Warsaw, Poland
| |
Collapse
|
4
|
Zhao RY, Wei PJ, Sun X, Zhang DH, He QY, Liu J, Chang JL, Yang Y, Guo ZN. Role of lipocalin 2 in stroke. Neurobiol Dis 2023; 179:106044. [PMID: 36804285 DOI: 10.1016/j.nbd.2023.106044] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 01/22/2023] [Accepted: 02/12/2023] [Indexed: 02/18/2023] Open
Abstract
Stroke is the second leading cause of death worldwide; however, the treatment choices available to neurologists are limited in clinical practice. Lipocalin 2 (LCN2) is a secreted protein, belonging to the lipocalin superfamily, with multiple biological functions in mediating innate immune response, inflammatory response, iron-homeostasis, cell migration and differentiation, energy metabolism, and other processes in the body. LCN2 is expressed at low levels in the brain under normal physiological conditions, but its expression is significantly up-regulated in multiple acute stimulations and chronic pathologies. An up-regulation of LCN2 has been found in the blood/cerebrospinal fluid of patients with ischemic/hemorrhagic stroke, and could serve as a potential biomarker for the prediction of the severity of acute stroke. LCN2 activates reactive astrocytes and microglia, promotes neutrophil infiltration, amplifies post-stroke inflammation, promotes blood-brain barrier disruption, white matter injury, and neuronal death. Moreover, LCN2 is involved in brain injury induced by thrombin and erythrocyte lysates, as well as microvascular thrombosis after hemorrhage. In this paper, we review the role of LCN2 in the pathological processes of ischemic stroke; intracerebral hemorrhage; subarachnoid hemorrhage; and stroke-related brain diseases, such as vascular dementia and post-stroke depression, and their underlying mechanisms. We hope that this review will help elucidate the value of LCN2 as a therapeutic target in stroke.
Collapse
Affiliation(s)
- Ruo-Yu Zhao
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Peng-Ju Wei
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Sun
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Dian-Hui Zhang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Qian-Yan He
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Jie Liu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China
| | - Jun-Lei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yi Yang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China; Neuroscience Research Center, the First Hospital of Jilin University, Chang Chun, China; Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China.
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Chang Chun, China; Neuroscience Research Center, the First Hospital of Jilin University, Chang Chun, China; Jilin Provincial Key Laboratory of Cerebrovascular Disease, Changchun, China.
| |
Collapse
|
5
|
Identification of Malignant Cell Populations Associated with Poor Prognosis in High-Grade Serous Ovarian Cancer Using Single-Cell RNA Sequencing. Cancers (Basel) 2022; 14:cancers14153580. [PMID: 35892844 PMCID: PMC9331511 DOI: 10.3390/cancers14153580] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Ovarian cancer has a high recurrence rate (~75%), and tumor heterogeneity is associated with such tumor recurrence. However, it is still poorly understood in ovarian cancer. To reveal tumor heterogeneity, we performed single-cell RNA sequencing (RNA-seq) of serous ovarian cancer cells from four different patients: two with primary carcinoma, one with recurrent carcinoma, and one with carcinoma treated with interval debulking surgery. As a result, we found two malignant tumor cell subtypes associated with poor prognosis. One malignant population included the earliest cancer cells and cancer stem-like cells. SLC3A1 and PEG10 were identified as the marker genes of cancer-initiating cells. The other malignant population expressing CA125 (MUC16), the most common biomarker for ovarian cancer, is associated with a decrease in the number of tumor-infiltrating cytotoxic T lymphocytes (CTLs). Our findings will offer new markers for diagnosis and choosing treatments targeting the malignant populations in ovarian cancer. Abstract To reveal tumor heterogeneity in ovarian cancer, we performed single-cell RNA sequencing (RNA-seq). We obtained The Cancer Genome Atlas (TCGA) survival data and TCGA gene expression data for a Kaplan–Meier plot showing the association of each tumor population with poor prognosis. As a result, we found two malignant tumor cell subtypes associated with poor prognosis. Next, we performed trajectory analysis using scVelo and Monocle3 and cell–cell interaction analysis using CellphoneDB. We found that one malignant population included the earliest cancer cells and cancer stem-like cells. Furthermore, we identified SLC3A1 and PEG10 as the marker genes of cancer-initiating cells. The other malignant population expressing CA125 (MUC16) is associated with a decrease in the number of tumor-infiltrating cytotoxic T lymphocytes (CTLs). Moreover, cell–cell interaction analysis implied that interactions mediated by LGALS9 and GAS6, expressed by this malignant population, caused the CTL suppression. The results of this study suggest that two tumor cell populations, including a cancer-initiating cell population and a population expressing CA125, survive the initial treatment and suppress antitumor immunity, respectively, and are associated with poor prognosis. Our findings offer a new understanding of ovarian cancer heterogeneity and will aid in the development of diagnostic tools and treatments.
Collapse
|
6
|
Astroglial and oligodendroglial markers in the cuprizone animal model for de- and remyelination. Histochem Cell Biol 2022; 158:15-38. [PMID: 35380252 PMCID: PMC9246805 DOI: 10.1007/s00418-022-02096-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2022] [Indexed: 01/08/2023]
Abstract
Myelin loss with consecutive axon degeneration and impaired remyelination are the underlying causes of progressive disease in patients with multiple sclerosis. Astrocytes are suggested to play a major role in these processes. The unmasking of distinct astrocyte identities in health and disease would help to understand the pathophysiological mechanisms in which astrocytes are involved. However, the number of specific astrocyte markers is limited. Therefore, we performed immunohistochemical studies and analyzed various markers including GFAP, vimentin, S100B, ALDH1L1, and LCN2 during de- and remyelination using the toxic murine cuprizone animal model. Applying this animal model, we were able to confirm overlapping expression of vimentin and GFAP and highlighted the potential of ALDH1L1 as a pan-astrocytic marker, in agreement with previous data. Only a small population of GFAP-positive astrocytes in the corpus callosum highly up-regulated LCN2 at the peak of demyelination and S100B expression was found in a subset of oligodendroglia as well, thus S100B turned out to have a limited use as a particular astroglial marker. Additionally, numerous GFAP-positive astrocytes in the lateral corpus callosum did not express S100B, further strengthening findings of heterogeneity in the astrocytic population. In conclusion, our results acknowledged that GFAP, vimentin, LCN2, and ALDH1L1 serve as reliable marker to identify activated astrocytes during cuprizone-induced de- and remyelination. Moreover, there were clear regional and temporal differences in protein and mRNA expression levels and patterns of the studied markers, generally between gray and white matter structures.
Collapse
|
7
|
Ren K, Xia Y. Lipocalin 2 Participates in the Epidermal Differentiation and Inflammatory Processes of Psoriasis. J Inflamm Res 2022; 15:2157-2166. [PMID: 35386225 PMCID: PMC8979418 DOI: 10.2147/jir.s358492] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/22/2022] [Indexed: 12/16/2022] Open
Abstract
As a multifunctional cytokine, lipocalin 2 is weakly expressed in skin and serum under normal conditions. However, it is over-expressed by neutrophils and keratinocytes in the skin lesions and sera in several skin diseases. Recent studies demonstrated that lipocalin 2 participates in the pathogenesis of psoriasis by exerting versatile effects on skin resident cells and infiltrating immune cells. Lipocalin 2 inhibits the synthesis of keratin, involucrin, and loricrin in keratinocytes, leading to epidermal parakeratosis via the Tcf7l1-lipocalin 2 signaling axis. It also recruits inflammatory cells such as T cells and neutrophils into skin lesions via the IL-23/IL17, p38-MAPK, and ERK-1/2 signaling pathways. Additionally, lipocalin 2 and other cytokines such as IL-17 have the synergetic effects on skin cells. The neutralization of lipocalin 2 or relevant cytokines can alleviate psoriasis, verifying that lipocalin 2 is an effective interfering target for psoriasis. In this review, we summarize the roles of lipocalin 2 in the processes of psoriatic inflammation and the promising therapeutic strategies based on lipocalin 2-related molecules.
Collapse
Affiliation(s)
- Kaixuan Ren
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
- Correspondence: Yumin Xia, Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, 157 Xiwu Road, Xi’an, 710004, People’s Republic of China, Tel/Fax +86-29-87679969, Email
| |
Collapse
|
8
|
Magnetic Resonance Imaging of Contrast-Induced Acute Renal Injury and Related Pathological Alterations In Vivo. Anal Cell Pathol (Amst) 2022; 2022:6984200. [PMID: 35256925 PMCID: PMC8898140 DOI: 10.1155/2022/6984200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/21/2022] [Accepted: 01/31/2022] [Indexed: 11/18/2022] Open
Abstract
Background. The definitive mechanisms of CI-AKI include contrast medium (CM) nephrotoxicity and CM disturbances in renal blood flow, but how the immune system responds to CM has rarely been mentioned in previous studies, and different cell death pathways have not been clearly distinguished. Aim. To confirm whether MRI detect early CI-AKI and to investigate whether immunity-related responses, pyroptosis, and mitophagy participate in contrast-induced acute renal injury (CI-AKI). Methods. C57BL/6 mice with CI-AKI were established by tail vein injection of iodixanol 320. Magnetic resonance imaging of 9.4 T scanner and microscopic appearance of renal H&E staining were tools to test the occurrence of CI-AKI at different times. Immunohistochemistry and NGAL were used to examine the immune responses in the kidneys with CI-AKI. Transmission electron microscopy and western blot methods were used to distinguish various cell death pathways in CI-AKI. Key Results. The densitometry of T2WI, DTI, and BOLD presents CI-AKI in a regular way. The microscopic appearance presents the strongest renal damage in CI-AKI mice that existed between 12 h (
) and 24 h (
) after contrast medium (CM) injection. Strong correlation may exist between MRI densitometry (T2WI, DTI, and BOLD) and pathology. Neutrophil and macrophage chemotaxis occurred in CI-AKI, and we observed that Ly6G was the strongest at 48 h (
). Pyroptosis (Nlrp3/caspase-1,
), mitophagy (BNIP/Nix,
), and apoptosis (Bax,
) occurred in CI-AKI. Conclusions. fMRI can detect early CI-AKI immediately after CM injection. NLRP3 inflammasomes are involved in CI-AKI, and mitophagy may play a role in mitigating kidney injury. The mitochondrion is one of the key organelles in the tubular epithelium implicated in CI-AKI.
Collapse
|
9
|
El-Sayes N, Vito A, Salem O, Workenhe ST, Wan Y, Mossman K. A Combination of Chemotherapy and Oncolytic Virotherapy Sensitizes Colorectal Adenocarcinoma to Immune Checkpoint Inhibitors in a cDC1-Dependent Manner. Int J Mol Sci 2022; 23:1754. [PMID: 35163675 PMCID: PMC8915181 DOI: 10.3390/ijms23031754] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/13/2022] [Accepted: 01/18/2022] [Indexed: 02/04/2023] Open
Abstract
Immune checkpoint therapy has shown great promise in the treatment of cancers with a high mutational burden, such as mismatch repair-deficient colorectal carcinoma (dMMR CRC). However, many patients fail to respond to immune checkpoint therapy. Using a mouse model of dMMR CRC, we demonstrated that tumors can be further sensitized to immune checkpoint therapy by using a combination of low-dose chemotherapy and oncolytic HSV-1. This combination induced the infiltration of CD8+ and CD4+ T cells into the tumor and the upregulation of gene signatures associated with the chemoattraction of myeloid cell subsets. When combined with immune checkpoint therapy, the combination promoted the infiltration of activated type 1 conventional dendritic cells (cDC1s) into the tumor. Furthermore, we found this combination strategy to be dependent on cDC1s, and its therapeutic efficacy to be abrogated in cDC1-deficient Batf3-/- mice. Thus, we demonstrated that the adjuvanticity of dMMR CRCs can be improved by combining low-dose chemotherapy and oncolytic HSV-1 in a cDC1-dependent manner.
Collapse
Affiliation(s)
- Nader El-Sayes
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada; (N.E.-S.); (O.S.); (Y.W.)
- Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Alyssa Vito
- Department of Clinical Translation, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada;
| | - Omar Salem
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada; (N.E.-S.); (O.S.); (Y.W.)
| | - Samuel Tekeste Workenhe
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Yonghong Wan
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada; (N.E.-S.); (O.S.); (Y.W.)
| | - Karen Mossman
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON L8S 4K1, Canada; (N.E.-S.); (O.S.); (Y.W.)
| |
Collapse
|
10
|
Martínez-Alberquilla I, Gasull X, Pérez-Luna P, Seco-Mera R, Ruiz-Alcocer J, Crooke A. Neutrophils and neutrophil extracellular trap components: Emerging biomarkers and therapeutic targets for age-related eye diseases. Ageing Res Rev 2022; 74:101553. [PMID: 34971794 DOI: 10.1016/j.arr.2021.101553] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/17/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Abstract
Age-related eye diseases, including dry eye, glaucoma, age-related macular degeneration, and diabetic retinopathy, represent a major global health issue based on their increasing prevalence and disabling action. Unraveling the molecular mechanisms underlying these diseases will provide novel opportunities to reduce the burden of age-related eye diseases and improve eye health, contributing to sustainable development goals achievement. The impairment of neutrophil extracellular traps formation/degradation processes seems to be one of these mechanisms. These traps formed by a meshwork of DNA and neutrophil cytosolic granule proteins may exacerbate the inflammatory response promoting chronic inflammation, a pivotal cause of age-related diseases. In this review, we describe current findings that suggest the role of neutrophils and their traps in the pathogenesis of the above-mentioned age-related eye diseases. Furthermore, we discuss why these cells and their constituents could be biomarkers and therapeutic targets for dry eye, glaucoma, age-related macular degeneration, and diabetic retinopathy. We also examine the therapeutic potential of some neutrophil function modulators and provide several recommendations for future research in age-related eye diseases.
Collapse
Affiliation(s)
- Irene Martínez-Alberquilla
- Department of Optometry and Vision, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain; Clinical and Experimental Eye Research Group, UCM 971009, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Xavier Gasull
- Neurophysiology Laboratory, Department of Biomedicine, Medical School, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Patricia Pérez-Luna
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Rubén Seco-Mera
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Javier Ruiz-Alcocer
- Department of Optometry and Vision, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain; Clinical and Experimental Eye Research Group, UCM 971009, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain
| | - Almudena Crooke
- Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain; Clinical and Experimental Eye Research Group, UCM 971009, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
11
|
Araos P, Amador CA. Neutrophil gelatinase-associated lipocalin as an immunomodulator in endocrine hypertension. Front Endocrinol (Lausanne) 2022; 13:1006790. [PMID: 36387895 PMCID: PMC9640732 DOI: 10.3389/fendo.2022.1006790] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/04/2022] [Indexed: 12/04/2022] Open
Abstract
In recent studies, primary aldosteronism (PA) has been reported as the most common etiology for secondary hypertension of endocrine origin, accounting for approximately 10% of cases. In PA, excess aldosterone production can lead to deleterious effects at the cardiovascular (CV) and renal levels by activating mineralocorticoid receptors, which involves an increase in pro-inflammatory and pro-fibrotic mediators. Among these mediators, neutrophil gelatinase-associated lipocalin (NGAL), a secretion glycoprotein belonging to the lipocalin superfamily, has been closely linked to CV and renal damage in several pathological conditions. Because NGAL can be detected in biofluids such as plasma and urine, it has been proposed as a damage biomarker for target tissues and has also been studied for its role in hypertension and associated with PA. NGAL is produced by many different cell types, can be carried on extracellular vesicles, and is modulated by microRNAs, which would support its use as a biomarker for endocrine hypertension due to PA. Over the last decade, studies have shown that NGAL is necessary for the development of aldosterone-induced hypertension and that is associated with end-organ damage. In addition, it has been proposed that some mechanisms are dependent on the activation of immune cells, such as dendritic cells and macrophages, where the release of specific cytokines (i.e., interleukin [IL]-23) or chemokines (i.e., CCL-5) induced by aldosterone would depend on NGAL. Subsequently, this activates the T helper (Th) lymphocytes, such as Th17 and Th2, resulting in CV and renal fibrosis due to the high aldosterone levels. Although the immune system has been closely associated with essential hypertension, its participation in endocrine hypertension has not been fully elucidated. This review discusses the link between NGAL and endocrine hypertension, particularly in the context of PA, and their possible regulators and mechanisms, with a focus on its role as an immunomodulator.
Collapse
Affiliation(s)
- Patricio Araos
- Laboratorio de Fisiopatología Renal, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Cristián A. Amador
- Laboratorio de Fisiopatología Renal, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- *Correspondence: Cristián A. Amador,
| |
Collapse
|
12
|
Bonnard B, Ibarrola J, Lima-Posada I, Fernández-Celis A, Durand M, Genty M, Lopez-Andreés N, Jaisser F. Neutrophil Gelatinase-Associated Lipocalin From Macrophages Plays a Critical Role in Renal Fibrosis Via the CCL5 (Chemokine Ligand 5)-Th2 Cells-IL4 (Interleukin 4) Pathway. Hypertension 2021; 79:352-364. [PMID: 34794340 DOI: 10.1161/hypertensionaha.121.17712] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
NGAL (neutrophil gelatinase-associated lipocalin; or lipocalin 2, Lcn2) is a novel mineralocorticoid target in the cardiovascular system. We showed that Lcn2 gene invalidation protects against proteinuria and renal injury upon mineralocorticoid excess and we hypothesized that NGAL produced from macrophages promotes the expression of chemoattractant molecules involved these renal lesions. The role of NGAL was analyzed using myeloid-specific (MΦ KO NGAL) Lcn2 knockout mice challenged with uni-nephrectomy, aldosterone, and salt (NAS) for 6 weeks. The role of the CCL5 (chemokine ligand 5) and IL4 (interleukin 4) in kidney fibrosis was studied by administration of the CCL5 receptor antagonist maraviroc or by injections of an anti-IL4 neutralizing antibody. In CTL mice, NAS increased the renal expression of extracellular matrix proteins, such as collagen I, αSMA, and fibronectin associated with interstitial fibrosis which were blunted in MΦ KO NGAL mice. The expression of CCL5 was blunted in sorted macrophages from MΦ KO NGAL mice challenged by NAS and in macrophages obtained from KO NGAL mice and challenged ex vivo with aldosterone and salt. The pharmacological blockade of the CCL5 receptor reduced renal fibrosis and the CD4+ Th cell infiltration induced by NAS. Neutralization of IL4 in NAS mice blunted kidney fibrosis and the overexpression of profibrotic proteins, such as collagen I, αSMA, and fibronectin. In conclusion, NGAL produced by macrophages plays a critical role in renal fibrosis and modulates the CCL5/IL4 pathway in mice exposed to mineralocorticoid excess.
Collapse
Affiliation(s)
- Benjamin Bonnard
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Jaime Ibarrola
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Ixchel Lima-Posada
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Manon Durand
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Marie Genty
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Natalia Lopez-Andreés
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Frédéric Jaisser
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.).,INSERM, Clinical Investigation Centre 1433, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, Nancy, France (F.J.)
| |
Collapse
|
13
|
Tang KH, Li S, Khodadadi-Jamayran A, Jen J, Han H, Guidry K, Chen T, Hao Y, Fedele C, Zebala JA, Maeda DY, Christensen JG, Olson P, Athanas A, Loomis CA, Tsirigos A, Wong KK, Neel BG. Combined Inhibition of SHP2 and CXCR1/2 Promotes Anti-Tumor T Cell Response in NSCLC. Cancer Discov 2021; 12:47-61. [PMID: 34353854 DOI: 10.1158/2159-8290.cd-21-0369] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 06/29/2021] [Accepted: 07/26/2021] [Indexed: 11/16/2022]
Abstract
SHP2 inhibitors (SHP2i) alone and in various combinations are being tested in multiple tumors with over-activation of the RAS/ERK pathway. SHP2 plays critical roles in normal cell signaling; hence, SHP2is could influence the tumor microenvironment. We found that SHP2i treatment depleted alveolar and M2-like macrophages, induced tumor-intrinsic CCL5/CXCL10 secretion and promoted B and T lymphocyte infiltration in Kras- and Egfr-mutant non-small cell lung cancer (NSCLC). However, treatment also increased intratumor gMDSCs via tumor-intrinsic, NF-kB-dependent production of CXCR2 ligands. Other RAS/ERK pathway inhibitors also induced CXCR2 ligands and gMDSC influx in mice, and CXCR2 ligands were induced in tumors from patients on KRASG12C-inhibitor trials. Combined SHP2(SHP099)/CXCR1/2(SX682) inhibition depleted a specific cluster of S100a8/9high gMDSCs, generated Klrg1+ CD8+ effector T cells with a strong cytotoxic phenotype but expressing the checkpoint receptor NKG2A, and enhanced survival in Kras- and Egfr-mutant models. Our results argue for testing RAS/ERK pathway/CXCR1/2/NKG2A inhibitor combinations in NSCLC patients.
Collapse
Affiliation(s)
- Kwan Ho Tang
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York.
| | - Shuai Li
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Alireza Khodadadi-Jamayran
- Applied Bioinformatics Laboratories, Office of Science and Research, New York University Grossman School of Medicine, New York, New York
| | - Jayu Jen
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Han Han
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Kayla Guidry
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Ting Chen
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | - Yuan Hao
- Applied Bioinformatics Laboratories, Office of Science and Research, New York University Grossman School of Medicine, New York, New York
| | - Carmine Fedele
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York
| | | | | | | | - Peter Olson
- Mirati Therapeutics, Inc., San Diego, California
| | | | - Cynthia A Loomis
- Department of Pathology, New York University Grossman School of Medicine, New York, New York
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories, Office of Science and Research, New York University Grossman School of Medicine, New York, New York
- Department of Pathology, New York University Grossman School of Medicine, New York, New York
- Institute for Computational Medicine, New York University Grossman School of Medicine, New York, New York
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York.
| | - Benjamin G Neel
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, New York.
| |
Collapse
|
14
|
Regulation of the Nfkbiz Gene and Its Protein Product IkBζ in Animal Models of Sepsis and Endotoxic Shock. Infect Immun 2021; 89:IAI.00674-20. [PMID: 33431705 DOI: 10.1128/iai.00674-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022] Open
Abstract
Sepsis is a life-threatening condition that arises from a poorly regulated inflammatory response to pathogenic organisms. Current treatments are limited to antibiotics, fluid resuscitation, and other supportive therapies. New targets for monitoring disease progression and therapeutic interventions are therefore critically needed. We previously reported that lipocalin-2 (Lcn2), a bacteriostatic mediator with potent proapoptotic activities, was robustly induced in sepsis. Other studies showed that Lcn2 was a predictor of mortality in septic patients. However, how Lcn2 is regulated during sepsis is poorly understood. We evaluated how IkBζ, an inducer of Lcn2, was regulated in sepsis using both the cecal ligation and puncture (CLP) and endotoxemia (lipopolysaccharide [LPS]) animal models. We show that Nfkbiz, the gene encoding IkBζ, was rapidly stimulated but, unlike Lcn2, whose expression persists during sepsis, mRNA levels of Nfkbiz decline to near basal levels several hours after its induction. In contrast, we observed that IkBζ expression remained highly elevated in septic animals following CLP but not LPS, indicating the occurrence of a CLP-specific mechanism that extends IkBζ half-life. By using an inhibitor of IkBζ, we determined that the expression of Lcn2 was largely controlled by IkBζ. Altogether, these data indicate that the high IkBζ expression in tissues likely contributes to the elevated expression of Lcn2 in sepsis. Since IkBζ is also capable of promoting or repressing other inflammatory genes, it might exert a central role in sepsis.
Collapse
|
15
|
Treekitkarnmongkol W, Hassane M, Sinjab A, Chang K, Hara K, Rahal Z, Zhang J, Lu W, Sivakumar S, McDowell TL, Kantrowitz J, Zhou J, Lang W, Xu L, Ochieng JK, Nunomura-Nakamura S, Deng S, Behrens C, Raso MG, Fukuoka J, Reuben A, Ostrin EJ, Parra E, Solis LM, Spira AE, McAllister F, Cascone T, Wistuba II, Moghaddam SJ, Scheet PA, Fujimoto J, Kadara H. Augmented Lipocalin-2 Is Associated with Chronic Obstructive Pulmonary Disease and Counteracts Lung Adenocarcinoma Development. Am J Respir Crit Care Med 2021; 203:90-101. [PMID: 32730093 DOI: 10.1164/rccm.202004-1079oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Rationale: Early pathogenesis of lung adenocarcinoma (LUAD) remains largely unknown. We found that, relative to wild-type littermates, the innate immunomodulator Lcn2 (lipocalin-2) was increased in normal airways from mice with knockout of the airway lineage gene Gprc5a (Gprc5a-/-) and that are prone to developing inflammation and LUAD. Yet, the role of LCN2 in lung inflammation and LUAD is poorly understood.Objectives: Delineate the role of Lcn2 induction in LUAD pathogenesis.Methods: Normal airway brushings, uninvolved lung tissues, and tumors from Gprc5a-/- mice before and after tobacco carcinogen exposure were analyzed by RNA sequencing. LCN2 mRNA was analyzed in public and in-house data sets of LUAD, lung squamous cancer (LUSC), chronic obstructive pulmonary disease (COPD), and LUAD/LUSC with COPD. LCN2 protein was immunohistochemically analyzed in a tissue microarray of 510 tumors. Temporal lung tumor development, gene expression programs, and host immune responses were compared between Gprc5a-/- and Gprc5a-/-/Lcn2-/- littermates.Measurements and Main Results: Lcn2 was progressively elevated during LUAD development and positively correlated with proinflammatory cytokines and inflammation gene sets. LCN2 was distinctively elevated in human LUADs, but not in LUSCs, relative to normal lungs and was associated with COPD among smokers and patients with LUAD. Relative to Gprc5a-/- mice, Gprc5a-/-/Lcn2-/- littermates exhibited significantly increased lung tumor development concomitant with reduced T-cell abundance (CD4+) and richness, attenuated antitumor immune gene programs, and increased immune cell expression of protumor inflammatory cytokines.Conclusions: Augmented LCN2 expression is a molecular feature of COPD-associated LUAD and counteracts LUAD development in vivo by maintaining antitumor immunity.
Collapse
Affiliation(s)
| | - Maya Hassane
- Department of Biochemistry and Molecular Genetics and
| | | | | | - Kieko Hara
- Department of Translational Molecular Pathology
| | - Zahraa Rahal
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jiexin Zhang
- Department of Bioinformatics and Computer Biology
| | - Wei Lu
- Department of Translational Molecular Pathology
| | | | - Tina L McDowell
- Department of Translational Molecular Pathology.,Department of Epidemiology
| | - Jacob Kantrowitz
- Section of Computational Biomedicine, School of Medicine, Boston University, Boston, Massachusetts; and
| | | | - Wenhua Lang
- Department of Translational Molecular Pathology
| | - Li Xu
- Department of Translational Molecular Pathology
| | | | | | | | | | | | - Junya Fukuoka
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | | | | | - Edwin Parra
- Department of Translational Molecular Pathology
| | | | - Avrum E Spira
- Section of Computational Biomedicine, School of Medicine, Boston University, Boston, Massachusetts; and
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tina Cascone
- Department of Thoracic Head and Neck Medical Oncology
| | | | | | | | | | | |
Collapse
|
16
|
Antimicrobial peptides: bridging innate and adaptive immunity in the pathogenesis of psoriasis. Chin Med J (Engl) 2020; 133:2966-2975. [PMID: 33237697 PMCID: PMC7752697 DOI: 10.1097/cm9.0000000000001240] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial peptides (AMPs) are small molecules produced by a myriad of cells and play important roles not only in protecting against infections and sustaining skin barrier homeostasis but also in contributing to immune dysregulation under pathological conditions. Recently, increasing evidence has indicated that AMPs, including cathelicidin (LL-37), human β-defensins, S100 proteins, lipocalin 2, and RNase 7, are highly expressed in psoriatic skin lesions. These peptides broadly regulate immunity by interacting with various immune cells and linking innate and adaptive immune responses during the progression of psoriasis. In this review, we summarize the recent findings regarding AMPs in the pathogenesis of psoriasis with a main focus on their immunomodulatory abilities.
Collapse
|
17
|
Chen W, Li W, Zhang Z, Tang X, Wu S, Yao G, Li K, Wang D, Xu Y, Feng R, Duan X, Fan X, Lu L, Chen W, Li C, Sun L. Lipocalin-2 Exacerbates Lupus Nephritis by Promoting Th1 Cell Differentiation. J Am Soc Nephrol 2020; 31:2263-2277. [PMID: 32646856 DOI: 10.1681/asn.2019090937] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 05/24/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Lipocalin-2 (LCN2) is an indicator of the severity of lupus nephritis (LN) and plays a pivotal role in immune responses, but it is not known if its effect on LN pathogenesis derives from regulating the immune imbalance of T lymphocyte subsets. METHODS The expression of LCN2 in T cells and kidneys was assessed in renal biopsies from patients with LN. We investigated the relationship between LCN2 levels and development of LN and systemic illness by injecting anti-LCN2 antibodies into MRL/lpr mice and analyzing pristane-treated LCN2 -/- mice. RESULTS LCN2 is highly expressed in CD4+ T cells and in renal tissues, and is associated with severe renal damage in patients with LN and in mice with experimental lupus. LCN2 promotes IFN-γ overexpression in CD4+ T cells through the IL-12/STAT4 pathway in an autocrine or paracrine manner. Both neutralization of LCN2 in MRL/lpr mice and genetic depletion of LCN2 in pristane-induced lupus mice greatly ameliorate nephritis. The frequency and number of splenic and renal Th1 cells decrease in proportion to LN disease activity. Conversely, administration of LCN2 exacerbates the disease with significantly higher renal activity scores and increased numbers of Th1 cells. CONCLUSIONS LCN2 plays a crucial role in Th1 cell differentiation, and may present a potential therapeutic target for LN.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Wenchao Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Zhuoya Zhang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Shufang Wu
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Kang Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Yuemei Xu
- Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Ruihai Feng
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Xiaoxiao Duan
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China
| | - Xiangshan Fan
- Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Liwei Lu
- Department of Pathology, Center of Infection and Immunology, University of Hong Kong, Hong Kong, China
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Chaojun Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China .,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, China .,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| |
Collapse
|
18
|
Dendritic cells are crucial for cardiovascular remodeling and modulate neutrophil gelatinase-associated lipocalin expression upon mineralocorticoid receptor activation. J Hypertens 2020; 37:1482-1492. [PMID: 31033725 DOI: 10.1097/hjh.0000000000002067] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Adaptive immunity is crucial in cardiovascular and renal inflammation/fibrosis upon hyperactivation of mineralocorticoid receptor. We have previously demonstrated that dendritic cells can respond to mineralocorticoid receptor activation, and the neutrophil gelatinase-associated lipocalin (NGAL) in dendritic cells is highly increased during aldosterone (Aldo)/mineralocorticoid receptor-dependent cardiovascular damage. However, the interrelationship among dendritic cells, target organs inflammation/fibrosis induced by mineralocorticoid receptor, and NGAL-dependence remains unknown. OBJECTIVE We studied the role of dendritic cells in mineralocorticoid receptor-dependent tissue remodeling and whether NGAL can modulate the inflammatory response of dendritic cells after mineralocorticoid receptor activation. METHODS Cardiovascular and renal remodeling induced by Aldo and high-salt diet [nephrectomy-Aldo-salt (NAS) model] were analyzed in CD11c.DOG mice, a model which allows dendritic cells ablation by using diphtheria toxin. In addition, in-vitro studies in NGAL-knock out dendritic cells were performed to determine the immunomodulatory role of NGAL upon Aldo treatment. RESULTS The ablation of dendritic cells prevented the development of cardiac hypertrophy, perivascular fibrosis, and the overexpression of NGAL, brain natriuretic peptide, and two profibrotic factors induced by NAS: collagen 1A1 and connective tissue growth factor. We determined that dendritic cells were not required to prevent renal hypertrophy/fibrosis induced by NAS. Between different immune cells analyzed, we observed that NGAL abundance was higher in antigen-presenting cells, while in-vitro studies showed that mineralocorticoid receptor stimulation in dendritic cells favored NGAL and IL-23 expression (p19 and p40 subunits), which are involved in the development of fibrosis and the Th17-driven response, respectively. CONCLUSION NGAL produced by dendritic cells may play a pivotal role in the activation of adaptive immunity that leads to cardiovascular fibrosis during mineralocorticoids excess.
Collapse
|
19
|
Schreiber A, Rousselle A, Klocke J, Bachmann S, Popovic S, Bontscho J, Schmidt-Ott KM, Siffrin V, Jerke U, Ashraf MI, Panzer U, Kettritz R. Neutrophil Gelatinase-Associated Lipocalin Protects from ANCA-Induced GN by Inhibiting T H17 Immunity. J Am Soc Nephrol 2020; 31:1569-1584. [PMID: 32487561 DOI: 10.1681/asn.2019090879] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 04/14/2020] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Neutrophil gelatinase-associated lipocalin (NGAL) is a diagnostic marker of intrinsic kidney injury produced by damaged renal cells and by neutrophils. ANCA-associated vasculitis features necrotizing crescentic GN (NCGN), and ANCA-activated neutrophils contribute to NCGN. Whether NGAL plays a mechanistic role in ANCA-associated vasculitis is unknown. METHODS We measured NGAL in patients with ANCA-associated vasculitis and mice with anti-myeloperoxidase (anti-MPO) antibody-induced NCGN. We compared kidney histology, neutrophil functions, T cell proliferation and polarization, renal infiltrating cells, and cytokines in wild-type and NGAL-deficient chimeric mice with anti-MPO antibody-induced NCGN. To assess the role of TH17 immunity, we transplanted irradiated MPO-immunized MPO-deficient mice with bone marrow from either wild-type or NGAL-deficient mice; we also transplanted irradiated MPO-immunized MPO/IL-17A double-deficient mice with bone marrow from either IL-17A-deficient or NGAL/IL-17A double-deficient mice. RESULTS Mice and patients with active ANCA-associated vasculitis demonstrated strongly increased serum and urinary NGAL levels. ANCA-stimulated neutrophils released NGAL. Mice with NGAL-deficient bone marrow developed worsened MPO-ANCA-induced NCGN. Intrinsic neutrophil functions were similar in NGAL-deficient and wild-type neutrophils, whereas T cell immunity was increased in chimeric mice with NGAL-deficient neutrophils with more renal infiltrating TH17 cells. NGAL-expressing neutrophils and CD3+ T cells were in close proximity in kidney and spleen. CD4+ T cells showed no intrinsic difference in proliferation and polarization in vitro, whereas iron siderophore-loaded NGAL suppressed TH17 polarization. We found significantly attenuated NCGN in IL-17A-deficient chimeras compared with MPO-deficient mice receiving wild-type bone marrow, as well as in NGAL/IL-17A-deficient chimeras compared with NGAL-deficient chimeras. CONCLUSIONS Our findings support that bone marrow-derived, presumably neutrophil, NGAL protects from ANCA-induced NCGN by downregulating TH17 immunity.
Collapse
Affiliation(s)
- Adrian Schreiber
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité - Berlin University of Medicine, Corporate Member of Free University of Berlin, Humboldt University of Berlin, Berlin Institute of Health, Berlin, Germany .,Nephrology and Medical Intensive Care Medicine, Charité - Berlin University of Medicine, Berlin, Germany
| | - Anthony Rousselle
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité - Berlin University of Medicine, Corporate Member of Free University of Berlin, Humboldt University of Berlin, Berlin Institute of Health, Berlin, Germany
| | - Jan Klocke
- Nephrology and Medical Intensive Care Medicine, Charité - Berlin University of Medicine, Berlin, Germany
| | - Sebastian Bachmann
- Institute of Vegetative Anatomy, Charité - Berlin University of Medicine, Berlin, Germany
| | - Suncica Popovic
- Institute of Vegetative Anatomy, Charité - Berlin University of Medicine, Berlin, Germany
| | - Julia Bontscho
- Nephrology and Medical Intensive Care Medicine, Charité - Berlin University of Medicine, Berlin, Germany
| | - Kai M Schmidt-Ott
- Nephrology and Medical Intensive Care Medicine, Charité - Berlin University of Medicine, Berlin, Germany
| | - Volker Siffrin
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité - Berlin University of Medicine, Corporate Member of Free University of Berlin, Humboldt University of Berlin, Berlin Institute of Health, Berlin, Germany
| | - Uwe Jerke
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité - Berlin University of Medicine, Corporate Member of Free University of Berlin, Humboldt University of Berlin, Berlin Institute of Health, Berlin, Germany
| | - Muhammad Imtiaz Ashraf
- Department of Surgery, Campus Charité Mitte I Campus Virchow Klinikum, Charité - Berlin University of Medicine, Berlin, Germany
| | - Ulf Panzer
- III. Medical Clinic, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ralph Kettritz
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité - Berlin University of Medicine, Corporate Member of Free University of Berlin, Humboldt University of Berlin, Berlin Institute of Health, Berlin, Germany.,Nephrology and Medical Intensive Care Medicine, Charité - Berlin University of Medicine, Berlin, Germany
| |
Collapse
|
20
|
Emmer A, Abobarin-Adeagbo A, Posa A, Jordan B, Delank KS, Staege MS, Surov A, Zierz S, Kornhuber ME. Myositis in Lewis rats induced by the superantigen Staphylococcal enterotoxin A. Mol Biol Rep 2019; 46:4085-4094. [DOI: 10.1007/s11033-019-04858-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/03/2019] [Indexed: 12/18/2022]
|
21
|
Gorini S, Marzolla V, Mammi C, Armani A, Caprio M. Mineralocorticoid Receptor and Aldosterone-Related Biomarkers of End-Organ Damage in Cardiometabolic Disease. Biomolecules 2018; 8:biom8030096. [PMID: 30231508 PMCID: PMC6165349 DOI: 10.3390/biom8030096] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 12/11/2022] Open
Abstract
The mineralocorticoid receptor (MR) was first identified as a blood pressure regulator, modulating renal sodium handling in response to its principal ligand aldosterone. The mineralocorticoid receptor is also expressed in many tissues other than the kidney, such as adipose tissue, heart and vasculature. Recent studies have shown that MR plays a relevant role in the control of cardiovascular and metabolic function, as well as in adipogenesis. Dysregulation of aldosterone/MR signaling represents an important cause of disease as high plasma levels of aldosterone are associated with hypertension, obesity and increased cardiovascular risk. Aldosterone displays powerful vascular effects and acts as a potent pro-fibrotic agent in cardiovascular remodeling. Mineralocorticoid receptor activation regulates genes involved in vascular and cardiac fibrosis, calcification and inflammation. This review focuses on the role of novel potential biomarkers related to aldosterone/MR system that could help identify cardiovascular and metabolic detrimental conditions, as a result of altered MR activation. Specifically, we discuss: (1) how MR signaling regulates the number and function of different subpopulations of circulating and intra-tissue immune cells; (2) the role of aldosterone/MR system in mediating cardiometabolic diseases induced by obesity; and (3) the role of several MR downstream molecules as novel potential biomarkers of cardiometabolic diseases, end-organ damage and rehabilitation outcome.
Collapse
Affiliation(s)
- Stefania Gorini
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy.
| | - Vincenzo Marzolla
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy.
| | - Caterina Mammi
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy.
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy.
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy.
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy.
| |
Collapse
|
22
|
More than a simple biomarker: the role of NGAL in cardiovascular and renal diseases. Clin Sci (Lond) 2018; 132:909-923. [PMID: 29739822 DOI: 10.1042/cs20171592] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/05/2018] [Accepted: 04/04/2018] [Indexed: 12/15/2022]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) is a small circulating protein that is highly modulated in a wide variety of pathological situations, making it a useful biomarker of various disease states. It is one of the best markers of acute kidney injury, as it is rapidly released after tubular damage. However, a growing body of evidence highlights an important role for NGAL beyond that of a biomarker of renal dysfunction. Indeed, numerous studies have demonstrated a role for NGAL in both cardiovascular and renal diseases. In the present review, we summarize current knowledge concerning the involvement of NGAL in cardiovascular and renal diseases and discuss the various mechanisms underlying its pathological implications.
Collapse
|
23
|
Stoffel E, Maier H, Riedl E, Brüggen MC, Reininger B, Schaschinger M, Bangert C, Guenova E, Stingl G, Brunner PM. Analysis of anti-tumour necrosis factor-induced skin lesions reveals strong T helper 1 activation with some distinct immunological characteristics. Br J Dermatol 2018; 178:1151-1162. [PMID: 29143979 DOI: 10.1111/bjd.16126] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Psoriasiform and eczematous eruptions are the most common dermatological adverse reactions linked to anti-tumour necrosis factor (TNF)-α therapy. Yet, a detailed characterization of their immune phenotype is lacking. OBJECTIVES To characterize anti-TNF-α-induced inflammatory skin lesions at a histopathological, cellular and molecular level, compared with psoriasis, eczema (atopic dermatitis) and healthy control skin. METHODS Histopathological evaluation, gene expression (quantitative real-time polymerase chain reaction) and computer-assisted immunohistological studies (TissueFAXS) were performed on 19 skin biopsies from patients with inflammatory bowel disease (n = 17) and rheumatoid arthritis (n = 2) with new-onset inflammatory skin lesions during anti-TNF-α-therapy. RESULTS Although most biopsies showed a psoriasiform and/or spongiotic (eczematous) histopathological architecture, these lesions were inconsistent with either psoriasis or eczema on a molecular level using an established chemokine (C-C motif) ligand 27/inducible nitric oxide synthase classifier. Despite some differences in immune skewing depending on the specific histopathological reaction pattern, all anti-TNF-α-induced lesions showed strong interferon (IFN)-γ activation, at higher levels than in psoriasis or eczema. IFN-γ was most likely produced by CD3/CD4/Tbet-positive T helper 1 lymphocytes. CONCLUSIONS New-onset anti-TNF-α-induced eruptions previously classified as psoriasis or spongiotic dermatitis (eczema) exhibit a molecular profile that is different from either of these disorders.
Collapse
Affiliation(s)
- E Stoffel
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria.,Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - H Maier
- Department of Dermatology, Division of General Dermatology, Medical University of Vienna, Vienna, Austria
| | - E Riedl
- Department of Dermatology, Division of General Dermatology, Medical University of Vienna, Vienna, Austria
| | - M-C Brüggen
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - B Reininger
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - M Schaschinger
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - C Bangert
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - E Guenova
- Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - G Stingl
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria
| | - P M Brunner
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, Medical University of Vienna, Vienna, Austria.,Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, U.S.A
| |
Collapse
|
24
|
de Aragão-França LS, Aragão-França LS, Rocha VCJ, Rocha VCJ, Cronemberger-Andrade A, da Costa FHB, Costa FHB, Vasconcelos JF, Vasconcelos JF, Athanazio DA, Silva DN, Santos ES, Santos ES, Meira CS, Araujo CF, Araújo CF, Cerqueira JV, Cardillo F, Alcântara-Neves NM, Soares MBP, Pontes de Carvalho LC, Pontes-de-Carvalho LC. Tolerogenic Dendritic Cells Reduce Airway Inflammation in a Model of Dust Mite Triggered Allergic Inflammation. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:406-419. [PMID: 29949837 PMCID: PMC6021587 DOI: 10.4168/aair.2018.10.4.406] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/02/2018] [Accepted: 02/14/2018] [Indexed: 12/29/2022]
Abstract
PURPOSE The use of tolerogenic dendritic cells (TolDCs) to control exacerbated immune responses may be a prophylactic and therapeutic option for application in autoimmune and allergic conditions. The objective of this work was to evaluate the effects of TolDC administration in a mouse model of allergic airway inflammation caused by mite extract. METHODS Mouse bone marrow-derived TolDCs were induced by incubation with granulocyte-macrophage colony-stimulating factor (GM-CSF) and dexamethasone, and then characterized by flow cytometry and cytokine production by enzyme-linked immunosorbent assay (ELISA). For the in vivo model of Blomia tropicalis-induced allergy, mice transplanted with antigen-pulsed TolDCs were sensitized intraperitoneally with B. tropicalis mite extract (BtE) adsorbed to aluminium hydroxide. After challenge by nasal administration of BtE, bronchoalveolar lavage fluid (BALF), lungs, spleen and serum were collected for analysis. RESULTS Induction of TolDCs was efficiently achieved as shown by low expression of major histocompatibility complex (MHC) II, programmed death-ligand (PD-L) 2 and pro-inflammatory cytokine production, and up-regulation of interleukin (IL)-10, upon LPS stimulation in vitro. Transplantation of 1 or 2 doses of BtE-pulsed TolDCs reduced the number of inflammatory cells in BALF and lungs as well as mucus deposition. Moreover, compared to saline-injected controls, TolDC-treated mice showed lower serum levels of anti-BtE immunoglobulin E (IgE) antibodies as well as reduced Gata3 and IL-4 gene expression in the lungs and decreased IFN-γ levels in the supernatant of splenocyte cultures Transplantation of TolDCs increased the percentage of the regulatory T cells in the spleen and the lungs. CONCLUSIONS Preventive treatment with TolDCs protects against dust mite-induced allergy in a mouse model, reinforcing the use of tolerogenic dendritic cells for the management of allergic conditions.
Collapse
Affiliation(s)
| | - Luciana S Aragão-França
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil.,Centro de Biotecnologia e Terapia Celular, Hospital São Rafael, Salvador, Bahia, Brazil
| | | | - Viviane C J Rocha
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
| | | | | | - F H B Costa
- Department of Diagnostics and Biomedical Sciences at The University of Texas Health Science Center, Houston, USA
| | | | - José Fernandes Vasconcelos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil.,Centro de Biotecnologia e Terapia Celular, Hospital São Rafael, Salvador, Bahia, Brazil
| | - Daniel Abensur Athanazio
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil.,Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | | | | | - E S Santos
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
| | - Cássio Santana Meira
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
| | | | - C F Araújo
- Hospital Universitário Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | | | - Fabíola Cardillo
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
| | | | - Milena Botelho Pereira Soares
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil.,Centro de Biotecnologia e Terapia Celular, Hospital São Rafael, Salvador, Bahia, Brazil.
| | | | | |
Collapse
|
25
|
Neutrophil Gelatinase-Associated Lipocalin from immune cells is mandatory for aldosterone-induced cardiac remodeling and inflammation. J Mol Cell Cardiol 2017; 115:32-38. [PMID: 29289651 DOI: 10.1016/j.yjmcc.2017.12.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/05/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
Immune system activation is involved in cardiovascular (CV) inflammation and fibrosis, following activation of the mineralocorticoid receptor (MR). We previously showed that Neutrophil Gelatinase-Associated Lipocalin (NGAL) is a novel target of MR signaling in CV tissue and plays a critical role in aldosterone/MR-dependent hypertension and fibrosis. We hypothesized that the production of NGAL by immune cells may play an important part in the mediation of these deleterious mineralocorticoid-induced effects. We analyzed the effect of aldosterone on immune cell recruitment and NGAL expression in vivo. We then studied the role of NGAL produced by immune cells in aldosterone-mediated cardiac inflammation and remodeling using mice depleted for NGAL in their immune cells by bone marrow transplantation and subjected to mineralocorticoid challenge NAS (Nephrectomy, Aldosterone 200μg/kg/day, Salt 1%). NAS treatment induced the recruitment of various immune cell populations to lymph nodes (granulocytes, B lymphocytes, activated CD8+ T lymphocytes) and the induction of NGAL expression in macrophages, dendritic cells, and PBMCs. Mice depleted for NGAL in their immune cells were protected against NAS-induced cardiac remodeling and inflammation. We conclude that NGAL produced by immune cells plays a pivotal role in cardiac damage under mineralocorticoid excess. Our data further stressed a pathogenic role of NGAL in cardiac damages, besides its relevance as a biomarker of renal injury.
Collapse
|
26
|
Spleen-derived lipocalin-2 in the portal vein regulates Kupffer cells activation and attenuates the development of liver fibrosis in mice. J Transl Med 2017; 97:890-902. [PMID: 28504685 DOI: 10.1038/labinvest.2017.44] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 03/25/2017] [Accepted: 03/31/2017] [Indexed: 12/16/2022] Open
Abstract
The liver has an immune tolerance against gut-derived products from the portal vein (PV). A disruption of the gut-liver axis leads to liver injury and fibrosis. The spleen is connected to the PV and regulates immune functions. However, possible splenic effects on liver fibrosis development are unclear. Lipocalin-2 (Lcn2) is an antimicrobial protein that regulates macrophage activation. To clarify the role of the spleen in liver fibrosis development, we induced liver fibrosis in mice after splenectomy, and investigated liver fibrosis development. Liver fibrosis resulted in significantly increased splenic Lcn2 levels, but all other measured cytokine levels were unchanged. Splenectomized mice showed enhanced liver fibrosis and inflammation accompanied by significantly decreased Lcn2 levels in PV. Lipopolysaccharide-stimulated primary Kupffer cells, resident liver macrophages, which were treated with recombinant Lcn2 (rLcn2) produced less tumor necrosis factor-α and Ccl2 and the activation of hepatic stellate cells, the effector cells for collagen production in the liver, was suppressed by co-culture with rLcn2-treated Kupffer cells. In addition, the involvement of gut-derived products in splenectomized mice was evaluated by gut sterilization. Interestingly, gut sterilization blocked the effect of splenectomy on liver fibrosis development. In conclusion, spleen deficiency accelerated liver fibrosis development and decreased PV Lcn2 levels. The mechanism of splenic protection against liver fibrosis development may involve the splenic Lcn2, triggered by gut-derived products that enter the liver through the PV, regulates Kupffer cells activated by the gut-liver axis. Thus, the splenic Lcn2 may have an important role in regulating the immune tolerance of the liver in liver fibrosis development.
Collapse
|
27
|
Leone DA, Peschel A, Brown M, Schachner H, Ball MJ, Gyuraszova M, Salzer-Muhar U, Fukuda M, Vizzardelli C, Bohle B, Rees AJ, Kain R. Surface LAMP-2 Is an Endocytic Receptor That Diverts Antigen Internalized by Human Dendritic Cells into Highly Immunogenic Exosomes. THE JOURNAL OF IMMUNOLOGY 2017; 199:531-546. [DOI: 10.4049/jimmunol.1601263] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 05/10/2017] [Indexed: 12/13/2022]
|
28
|
Ashraf MI, Schwelberger HG, Brendel KA, Feurle J, Andrassy J, Kotsch K, Regele H, Pratschke J, Maier HT, Aigner F. Exogenous Lipocalin 2 Ameliorates Acute Rejection in a Mouse Model of Renal Transplantation. Am J Transplant 2016; 16:808-20. [PMID: 26595644 PMCID: PMC4996417 DOI: 10.1111/ajt.13521] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 09/02/2015] [Accepted: 09/04/2015] [Indexed: 01/25/2023]
Abstract
Lipocalin 2 (Lcn2) is rapidly produced by damaged nephron epithelia and is one of the most promising new markers of renal injury, delayed graft function and acute allograft rejection (AR); however, the functional importance of Lcn2 in renal transplantation is largely unknown. To understand the role of Lcn2 in renal AR, kidneys from Balb/c mice were transplanted into C57Bl/6 mice and vice versa and analyzed for morphological and physiological outcomes of AR at posttransplantation days 3, 5, and 7. The allografts showed a steady increase in intensity of interstitial infiltration, tubulitis and periarterial aggregation of lymphocytes associated with a substantial elevation in serum levels of creatinine, urea and Lcn2. Perioperative administration of recombinant Lcn2:siderophore:Fe complex (rLcn2) to recipients resulted in functional and morphological amelioration of the allograft at day 7 almost as efficiently as daily immunosuppression with cyclosporine A (CsA). No significant differences were observed in various donor-recipient combinations (C57Bl/6 wild-type and Lcn2(-/-) , Balb/c donors and recipients). Histochemical analyses of the allografts showed reduced cell death in recipients treated with rLcn2 or CsA. These results demonstrate that Lcn2 plays an important role in reducing the extent of kidney AR and indicate the therapeutic potential of Lcn2 in transplantation.
Collapse
Affiliation(s)
- M. I. Ashraf
- Department of VisceralTransplant and Thoracic SurgeryMedical University InnsbruckInnsbruckAustria,Department for General, Visceral and Transplantation Surgery, Campus Virchow‐KlinikumCharité UniversitätsmedizinBerlinGermany
| | - H. G. Schwelberger
- Department of VisceralTransplant and Thoracic SurgeryMedical University InnsbruckInnsbruckAustria
| | - K. A. Brendel
- Institute of PathologyMedical University InnsbruckInnsbruckAustria
| | - J. Feurle
- Department of VisceralTransplant and Thoracic SurgeryMedical University InnsbruckInnsbruckAustria
| | - J. Andrassy
- Department of Surgery, Clinic GrosshadernLudwig‐Maximilian‐University MunichMunichGermany
| | - K. Kotsch
- Department of VisceralTransplant and Thoracic SurgeryMedical University InnsbruckInnsbruckAustria
| | - H. Regele
- Institute of PathologyMedical University InnsbruckInnsbruckAustria
| | - J. Pratschke
- Department of VisceralTransplant and Thoracic SurgeryMedical University InnsbruckInnsbruckAustria,Department for General, Visceral and Transplantation Surgery, Campus Virchow‐KlinikumCharité UniversitätsmedizinBerlinGermany
| | - H. T. Maier
- Department of VisceralTransplant and Thoracic SurgeryMedical University InnsbruckInnsbruckAustria
| | - F. Aigner
- Department of VisceralTransplant and Thoracic SurgeryMedical University InnsbruckInnsbruckAustria,Department for General, Visceral and Transplantation Surgery, Campus Virchow‐KlinikumCharité UniversitätsmedizinBerlinGermany
| |
Collapse
|
29
|
Zinkevičienė A, Kainov D, Lastauskienė E, Kvedarienė V, Bychkov D, Byrne M, Girkontaitė I. Serum Biomarkers of Allergic Contact Dermatitis: A Pilot Study. Int Arch Allergy Immunol 2016; 168:161-4. [PMID: 26790150 DOI: 10.1159/000442749] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/24/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Allergic contact dermatitis (ACD) is an inflammatory skin disease caused by repeated skin exposure to contact allergens. The goal of this pilot study was to identify inflammatory proteins which can serve as biomarkers for ACD. METHODS We measured levels of 102 cytokines, chemokines, and growth factors in the sera of 16 ACD patients during acute and remission phases, and 16 healthy volunteers. RESULTS Serum levels of adiponectin, chemokine (C-C motif) ligand 5 (CCL5), C-reactive protein (CRP), chitinase 3-like 1 (CHI3L1), complement factor D (CFD), endoglin, lipocalin-2, osteopontin, retinol-binding protein 4 (RBP4), and platelet factor 4 (PF4) were significantly higher, whereas levels of trefoil factor 3 (TFF3) were significantly lower, in ACD patients than in healthy controls. In ACD patients, serum levels of CCL5 were elevated, whereas levels of TFF3, soluble intercellular adhesion molecule-1 (sICAM-1), and platelet-derived growth factor (PDGF)-AB/BB were found to be lower during the remission phase of the disease. CONCLUSIONS Serum levels of adiponectin, CCL5, CRP, CHI3L1, CFD, endoglin, lipocalin-2, osteopontin, RBP4, PF4, and TFF3 might be exploited as biomarkers for ACD, whereas levels of CCL5, TFF3, sICAM-1, and PDGF-AB/BB might be exploited for evaluation of disease progression and efficacy of ACD treatment.
Collapse
Affiliation(s)
- Auksė Zinkevičienė
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | | | | | | | | | | | | |
Collapse
|