1
|
Olivier DW, Eksteen C, Plessis MD, de Jager L, Engelbrecht L, McGregor NW, Shridas P, de Beer FC, de Villiers WJS, Pretorius E, Engelbrecht AM. Inflammation and Tumor Progression: The Differential Impact of SAA in Breast Cancer Models. BIOLOGY 2024; 13:654. [PMID: 39336082 PMCID: PMC11429026 DOI: 10.3390/biology13090654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/30/2024]
Abstract
Background: Previous research has shown that the Serum Amyloid A (SAA) protein family is intricately involved in inflammatory signaling and various disease pathologies. We have previously demonstrated that SAA is associated with increased colitis disease severity and the promotion of tumorigenesis. However, the specific role of SAA proteins in breast cancer pathology remains unclear. Therefore, we investigated the role of systemic SAA1 and SAA2 (SAA1/2) in a triple-negative breast cancer mouse model. Methods: Syngeneic breast tumors were established in wild-type mice, and mice lacking the SAA1/2 (SAADKO). Subsequently, tumor volume was monitored, species survival determined, the inflammatory profiles of mice assessed with a multiplex assay, and tumor molecular biology and histology characterized with Western blotting and H&E histological staining. Results: WT tumor-bearing mice had increased levels of plasma SAA compared to wild-type control mice, while SAADKO control and tumor-bearing mice presented with lower levels of SAA in their plasma. SAADKO tumor-bearing mice also displayed significantly lower concentrations of systemic inflammatory markers. Tumors from SAADKO mice overall had lower levels of SAA compared to tumors from wild-type mice, decreased apoptosis and inflammasome signaling, and little to no tumor necrosis. Conclusions: We demonstrated that systemic SAA1/2 stimulates the activation of the NLRP3 inflammasome in breast tumors, leading to the production of pro-inflammatory cytokines. This, in turn, promoted apoptosis and tumor necrosis but did not significantly impact tumor growth or histological grading.
Collapse
Affiliation(s)
- Daniel Wilhelm Olivier
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, Western Cape, South Africa
| | - Carla Eksteen
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, Western Cape, South Africa
| | - Manisha du Plessis
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, Western Cape, South Africa
| | - Louis de Jager
- Division of Anatomical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch 7600, Western Cape, South Africa
- National Health Laboratory Service, Tygerberg Hospital, Cape Town 7505, Western Cape, South Africa
| | - Lize Engelbrecht
- Central Analytical Facilities, Fluorescence Microscopy Unit, Stellenbosch University, Stellenbosch 7600, Western Cape, South Africa
| | - Nathaniel Wade McGregor
- Department of Genetics, Faculty of Agrisciences, Stellenbosch University, Stellenbosch 7600, Western Cape, South Africa
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, Western Cape, South Africa
| | - Preetha Shridas
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Frederick C de Beer
- Department of Internal Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Willem J S de Villiers
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, Western Cape, South Africa
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, Western Cape, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, Western Cape, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, Western Cape, South Africa
- Department of Global Health, African Cancer Institute (ACI), Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch 7505, Western Cape, South Africa
| |
Collapse
|
2
|
Erickson MA, Mahankali AP. Interactions of Serum Amyloid A Proteins with the Blood-Brain Barrier: Implications for Central Nervous System Disease. Int J Mol Sci 2024; 25:6607. [PMID: 38928312 PMCID: PMC11204325 DOI: 10.3390/ijms25126607] [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: 04/15/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Serum amyloid A (SAA) proteins are highly conserved lipoproteins that are notoriously involved in the acute phase response and systemic amyloidosis, but their biological functions are incompletely understood. Recent work has shown that SAA proteins can enter the brain by crossing the intact blood-brain barrier (BBB), and that they can impair BBB functions. Once in the central nervous system (CNS), SAA proteins can have both protective and harmful effects, which have important implications for CNS disease. In this review of the thematic series on SAA, we discuss the existing literature that relates SAA to neuroinflammation and CNS disease, and the possible roles of the BBB in these relations.
Collapse
Affiliation(s)
- Michelle A. Erickson
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, WA 98108, USA
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA;
| | - Anvitha P. Mahankali
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA;
| |
Collapse
|
3
|
Al Yaseen M, Al Zahid H, Al-Haroon S. Amyloid Deposits in the Ligamentum Flavum Related to Lumbar Spinal Canal Stenosis and Lumbar Disc Degeneration. Cureus 2022; 14:e26221. [PMID: 35911371 PMCID: PMC9313137 DOI: 10.7759/cureus.26221] [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] [Accepted: 06/22/2022] [Indexed: 12/01/2022] Open
Abstract
Background: Amyloidosis is a protein conformational disorder, with distinctive features of accumulation of protein fibrils in different body tissues, causing a wide range of signs and symptoms. These amyloid fibrils are usually derived from about 30 different precursor proteins that have been identified. Although the most common tissue for their accumulation is cardiac, amyloidosis may appear in many other tissues, though rarely cause symptoms. One of these extracardiac tissues is the ligamentum flavum (LF). Participants and Methods: Patients with lumbar spinal canal stenosis or lumbar disc degeneration, scheduled for surgery, were included in the study. A total of 17 LF specimens were obtained from 16 patients with lumbar spinal stenosis (two specimens were taken from two consecutive stenotic levels belonging to one patient), and 11 LF specimens were obtained from 11 patients with lumbar disc degeneration. Tissue biopsy was taken from the LF at the affected level and was stained with special immunohistochemical stain to detect transthyretin (TTR)-related amyloidosis (ATTR). The diameters of the lumbar canal and the LF thickness were measured at the affected level by a radiologist. Results: This study includes 22 LF specimens. Male to female ratio was 5.4:4.6 with the mean age comparatively equal (M = 46 years for men and 48 years for women). The patients were divided into two groups: lumbar canal stenosis and lumbar disc degeneration. The result of the immunohistochemical stain towards TTR amyloid was positive in five out of 22 (22%) samples and all were from the stenosis group. The relationship of the LF thickness to the canal diameter in the positively stained stenosis group specimens was significant (p = 0.001). All the positive specimens were taken from levels L3−4 and L4−5. Conclusion: There was a significant relationship between LF thickness and canal stenosis in the positively stained specimens (towards TTR amyloid) of the stenosis group. However, the disc degeneration group showed no relationship between canal diameter and LF thickness; moreover, all the specimens of that group stained negative. Middle-age patients with canal stenosis proved to have a significant relationship to amyloid deposit LF hypertrophy.
Collapse
|
4
|
IL-17A Is the Critical Cytokine for Liver and Spleen Amyloidosis in Inflammatory Skin Disease. Int J Mol Sci 2022; 23:ijms23105726. [PMID: 35628531 PMCID: PMC9147816 DOI: 10.3390/ijms23105726] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/30/2022] Open
Abstract
Systemic amyloidosis is recognized as a serious complication of rheumatoid arthritis or inflammatory bowel disease, but also of inflammatory skin disease. However, the detailed molecular mechanism of amyloidosis associated with cutaneous inflammation remains unclear, and therapeutic approaches are limited. Here, we investigated the pathophysiology of amyloidosis secondary to cutaneous inflammation and the therapeutic effects of Janus kinase (JAK) inhibitors by examining a mouse model of spontaneous dermatitis (KCASP1Tg mice). Moreover, KCASP1Tg mice were crossed with interleukin-17A (IL-17A) knockout mice to generate IL-17A-/KCASP1Tg and examine the role of IL-17A in amyloidosis under cutaneous inflammation. KCASP1Tg mice showed severe amyloid deposition in the liver and spleen. Increased serum-neutral fat levels and decreased lymphocyte production were observed in the spleen. Overproduction of amyloidosis was partially ameliorated by the administration of JAK inhibitors and was further improved in IL-17A-/KCASP1Tg mice. IL-17A-producing cells included CD4, gamma delta, and CD8 T cells. In summary, our results from the analysis of a mouse model of dermatitis revealed that skin-derived inflammatory cytokines can induce amyloid deposition in the liver and spleen, and that the administration of JAK inhibitors and, even more, IL-17A ablation, reduced amyloidosis. This study demonstrates that active control of skin inflammation is essential to prevent internal organ amyloidosis.
Collapse
|
5
|
Janus Kinase Inhibitors Ameliorated Gastrointestinal Amyloidosis and Hypoalbuminemia in Persistent Dermatitis Mouse Model. Int J Mol Sci 2021; 23:ijms23010028. [PMID: 35008464 PMCID: PMC8744922 DOI: 10.3390/ijms23010028] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 12/28/2022] Open
Abstract
Malnutrition is not only regarded as a complication of rheumatoid arthritis and inflammatory bowel disease but also that of inflammatory skin disease; however, the mechanisms and efficacy of its treatment have not been elucidated. Using a mouse model of dermatitis, we investigated the pathophysiology of malnutrition in inflammatory skin conditions and efficacy of its treatment. We employed spontaneous skin inflammation mice models overexpressing human caspase-1 in the epidermal keratinocytes. Body weight, nutrition level, and α1-antitrypsin fecal concentration were measured. The gastrointestinal tract was histologically and functionally investigated. Fluorescein isothiocyanate (FITC)-dextran was forcibly fed on an empty stomach, and plasma FITC-dextran was measured. The treatment efficacy of antibodies against tumor necrosis factor-α (TNF-α) and interleukin (IL)-α/β as well as Janus kinase (JAK) inhibitors was investigated. Compared with wild-type littermates, the inflammatory skin mice models showed a lowered body weight, reduction of serum albumin level, amyloid deposition in the stomach, small intestine, and large intestine, and increased α1-antitrypsin fecal concentration. However, the plasma FITC-dextran was unchanged between the dermatitis models and wild-type littermates. The over-produced serum amyloid A1 in the liver was detected in the plasma in the dermatitis model. Antibodies against TNF-α and IL-α/β showed partial effects on amyloid deposition; however, JAK inhibitors improved gastrointestinal amyloidosis with the improvement of skin symptoms. Chronic dermatitis is closely related to secondary amyloidosis in the gastrointestinal tract, resulting in hypoalbuminemia. Therefore, active control of skin inflammation is essential for preventing gastrointestinal complications.
Collapse
|
6
|
Schmidtke L, Meineck M, Saurin S, Otten S, Gather F, Schrick K, Käfer R, Roth W, Kleinert H, Weinmann-Menke J, Pautz A. Knockout of the KH-Type Splicing Regulatory Protein Drives Glomerulonephritis in MRL-Fas lpr Mice. Cells 2021; 10:3167. [PMID: 34831390 PMCID: PMC8624031 DOI: 10.3390/cells10113167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/30/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
KH-type splicing regulatory protein (KSRP) is an RNA-binding protein that promotes mRNA decay and thereby negatively regulates cytokine expression at the post-transcriptional level. Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by dysregulated cytokine expression causing multiple organ manifestations; MRL-Faslpr mice are an established mouse model to study lupus disease pathogenesis. To investigate the impact of KSRP on lupus disease progression, we generated KSRP-deficient MRL-Faslpr mice (MRL-Faslpr/KSRP-/- mice). In line with the predicted role of KSRP as a negative regulator of cytokine expression, lupus nephritis was augmented in MRL-Faslpr/KSRP-/- mice. Increased infiltration of immune cells, especially of IFN-γ producing T cells and macrophages, driven by enhanced expression of T cell-attracting chemokines and adhesion molecules, seems to be responsible for worsened kidney morphology. Reduced expression of the anti-inflammatory interleukin-1 receptor antagonist may be another reason for severe inflammation. The increase of FoxP3+ T cells detected in the kidney seems unable to dampen the massive kidney inflammation. Interestingly, lymphadenopathy was reduced in MRL-Faslpr/KSRP-/- mice. Altogether, KSRP appears to have a complex role in immune regulation; however, it is clearly able to ameliorate lupus nephritis.
Collapse
Affiliation(s)
- Lisa Schmidtke
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Myriam Meineck
- First Medical Department, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany;
| | - Sabrina Saurin
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
- First Medical Department, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany;
| | - Svenja Otten
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Fabian Gather
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Katharina Schrick
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Rudolf Käfer
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Wilfried Roth
- Institute of Pathology, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany;
| | - Hartmut Kleinert
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| | - Julia Weinmann-Menke
- First Medical Department, University Medical Center, Johannes Gutenberg-University Mainz, 55131 Mainz, Germany;
| | - Andrea Pautz
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (L.S.); (S.S.); (S.O.); (F.G.); (K.S.); (R.K.); (H.K.)
| |
Collapse
|
7
|
Bao J, Ma Y, Ding M, Wang C, Du G, Zhou Y, Guo L, Kang H, Wang C, Gu B. Preliminary exploration on the serum biomarkers of bloodstream infection with carbapenem-resistant Klebsiella pneumoniae based on mass spectrometry. J Clin Lab Anal 2021; 35:e23915. [PMID: 34331328 PMCID: PMC8418493 DOI: 10.1002/jcla.23915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/06/2021] [Accepted: 07/11/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Carbapenem-resistant K. pneumoniae (CRKP) bloodstream infections (BSI) must be rapidly identified to improve patient survival rates. This study investigated a new mass spectrometry-based method for improving the identification of CRKP BSI and explored potential biomarkers that could differentiate CRKP BSI from sensitive. METHODS Mouse models of BSI were first established. MALDI-TOF MS was then used to profile serum peptides in CRKP BSI versus normal samples before applying BioExplorer software to establish a diagnostic model to distinguish CRKP from normal. The diagnostic value of the model was then tested against 32 clinical CRKP BSI and 27 healthy serum samples. Finally, the identities of the polypeptides used to establish the diagnostic model were determined by secondary mass spectrometry. RESULTS 107 peptide peaks were shared between the CRKP and normal groups, with 18 peaks found to be differentially expressed. Five highly expressed peptides in the CRKP group (m/z 1349.8, 2091.3, 2908.2, 4102.1, and 8129.5) were chosen to establish a diagnostic model. The accuracy, specificity and sensitivity of the model were determined as 79.66%, 81.48%, and 78.12%, respectively. Secondary mass spectrometry identified the Fibrinogen alpha chain (FGA), Inter-alpha-trypsin inhibitor heavy chain H4 (ITIH4) and Serum amyloid A-2 protein (SAA2) as the source of the 5 serum peptides. CONCLUSIONS We successfully established a serum peptide-based diagnostic model that distinguished clinical CRKP BSI samples from normal healthy controls. The application of MALDI-TOF MS to measure serum peptides, therefore, represents a promising approach for early BSI diagnosis of BSI, especially for multidrug-resistant bacteria where identification is urgent.
Collapse
Affiliation(s)
- Jinfeng Bao
- College of Medical TechnologyXuzhou Medical UniversityXuzhouChina
- Department of Clinical LaboratoryThe First Medical CentreThe PLA General HospitalBeijingChina
| | - Yating Ma
- Department of Clinical LaboratoryThe First Medical CentreThe PLA General HospitalBeijingChina
| | - Mengshan Ding
- College of Medical TechnologyXuzhou Medical UniversityXuzhouChina
- Department of Clinical LaboratoryThe First Medical CentreThe PLA General HospitalBeijingChina
| | - Chi Wang
- Department of Clinical LaboratoryThe First Medical CentreThe PLA General HospitalBeijingChina
| | - Gaofei Du
- College of Medical TechnologyXuzhou Medical UniversityXuzhouChina
| | - Yuan Zhou
- College of Medical TechnologyXuzhou Medical UniversityXuzhouChina
| | - Ling Guo
- Department of Clinical LaboratoryThe First Medical CentreThe PLA General HospitalBeijingChina
| | - Haiquan Kang
- College of Medical TechnologyXuzhou Medical UniversityXuzhouChina
| | - Chengbin Wang
- Department of Clinical LaboratoryThe First Medical CentreThe PLA General HospitalBeijingChina
| | - Bing Gu
- College of Medical TechnologyXuzhou Medical UniversityXuzhouChina
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| |
Collapse
|
8
|
Dimond A, Van de Pette M, Fisher AG. Illuminating Epigenetics and Inheritance in the Immune System with Bioluminescence. Trends Immunol 2020; 41:994-1005. [PMID: 33036908 DOI: 10.1016/j.it.2020.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 12/25/2022]
Abstract
The remarkable process of light emission by living organisms has fascinated mankind for thousands of years. A recent expansion in the repertoire of catalytic luciferase enzymes, coupled with the discovery of the genes and pathways that encode different luciferin substrates, means that bioluminescence imaging (BLI) is set to revolutionize longitudinal and dynamic studies of gene control within biomedicine, including the regulation of immune responses. In this review article, we summarize recent advances in bioluminescence-based imaging approaches that promise to enlighten our understanding of in vivo gene and epigenetic control within the immune system.
Collapse
Affiliation(s)
- Andrew Dimond
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Mathew Van de Pette
- Epigenetic Mechanisms of Toxicity, MRC Toxicology Unit, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Amanda G Fisher
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
9
|
Weinhage T, Wirth T, Schütz P, Becker P, Lueken A, Skryabin BV, Wittkowski H, Foell D. The Receptor for Advanced Glycation Endproducts (RAGE) Contributes to Severe Inflammatory Liver Injury in Mice. Front Immunol 2020; 11:1157. [PMID: 32670276 PMCID: PMC7326105 DOI: 10.3389/fimmu.2020.01157] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/11/2020] [Indexed: 12/24/2022] Open
Abstract
Background: The receptor for advanced glycation end products (RAGE) is a multiligand receptor involved in a number of processes and disorders. While it is known that RAGE-signaling can contribute to toxic liver damage and fibrosis, its role in acute inflammatory liver injury and septic multiorgan failure is yet undefined. We examined RAGE in lipopolysaccharide (LPS) induced acute liver injury of D-galN sensitized mice as a classical model for tumor necrosis factor alpha (TNF-α) dependent inflammatory organ damage. Methods: Mice (Rage–/– and C57BL/6) were intraperitoneally injected with D-galN (300 mg/kg) and LPS (10 μg/kg). Animals were monitored clinically, and cytokines, damage associated molecular pattern molecules (DAMPs) as well as liver enzymes were determined in serum. Liver histology, hepatic cytokines as well as RAGE mRNA expression were analyzed. Cellular activation and functionality were evaluated by flow cytometry both in bone marrow- and liver-derived cells. Results: Genetic deficiency of RAGE significantly reduced the mortality of mice exposed to LPS/D-galN. Hepatocyte damage markers were reduced in Rage–/– mice, and liver histopathology was less severe. Rage–/– mice produced less pro-inflammatory cytokines and DAMPs in serum and liver. While immune cell functions appeared normal, TNF-α production by hepatocytes was reduced in Rage–/– mice. Conclusions: We found that RAGE deletion attenuated the expression of pro-inflammatory cytokines and DAMPs in hepatocytes without affecting cellular immune functions in the LPS/D-galN model of murine liver injury. Our data highlight the importance of tissue-specific RAGE-signaling also in acute inflammatory liver stress contributing to sepsis and multiorgan failure.
Collapse
Affiliation(s)
- Toni Weinhage
- Department of Pediatric Rheumatology and Immunology, University of Münster, Münster, Germany
| | - Timo Wirth
- Department of Neurology With Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Paula Schütz
- Department of Pediatric Rheumatology and Immunology, University of Münster, Münster, Germany
| | - Philipp Becker
- Department of Pediatric Rheumatology and Immunology, University of Münster, Münster, Germany
| | - Aloys Lueken
- Department of Pediatric Rheumatology and Immunology, University of Münster, Münster, Germany
| | - Boris V Skryabin
- Core Facility of Transgenic Animal and Genetic Engineering Models (TRAM), University of Münster, Münster, Germany
| | - Helmut Wittkowski
- Department of Pediatric Rheumatology and Immunology, University of Münster, Münster, Germany
| | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University of Münster, Münster, Germany
| |
Collapse
|
10
|
Luan X, Wang H, Xiang Z, Ma Z, Zhao J, Feng Y, Shi Q, Yin J. Biomimicking Dual-Responsive Extracellular Matrix Restoring Extracellular Balance through the Na/K-ATPase Pathway. ACS APPLIED MATERIALS & INTERFACES 2019; 11:21258-21267. [PMID: 31117462 DOI: 10.1021/acsami.9b05420] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Biomedical implant mimicking the physiological extracellular matrix (ECM) is a new strategy to modulate the cell microenvironment to improve implant integrity and longevity. However, the biomimicking ECM suffers from low sensitivity to pathological change and low efficiency to restore the physiological state in vivo. To overcome these problems, reactive oxygen species (ROS) and K+ dual-responsive micro-/nanofibers that encapsulate ascorbic acid-2-glucoside (AA-2G) are fabricated on an elastomer substrate with electrospinning to mimic the ECM. The strategy is based on the fact that ROS and K+ dual responsiveness enhance the sensitivity of the ECM to pathological changes and delivery of AA-2G from the ECM to cell membrane promotes reactivating Na/K-ATPase and shifting cellular diseased conditions to the normal state. We demonstrate that the ROS and K+-responsive tripolymer of poly(ethylene glycol)diacrylate, 1,2-ethanedithiol, and 4-nitrobenzo-18-crown-6-ether (PEGDA-EDT-BCAm) are synthesized successfully; the ECM composed of acylated poly(caprolactone)/PEGDA-EDT-BCAm/AA-2G micro-/nanofibers is prepared through reactive electrospinning; the ECM is sensitive to ROS and K+ concentration in the microenvironment to release AA-2G, which targets the membrane to remove the excessive ROS and reactivate Na/K-ATPase; as a result, the ECM reduces oxidative stress and restores the extracellular physiological state both in vitro and in vivo. This work provides basic principles to design an implant that can adjust the extracellular microenvironment while avoiding pathogenicity to improve implant integrity and longevity in vivo.
Collapse
Affiliation(s)
- Xingkun Luan
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , Jilin 130022 , China
- Key Laboratory of Olefin Catalysis and Polymerization/Key Laboratory of Rubber-Plastics (QUST) of Shandong Provincial , Qingdao University of Science and Technology , Qingdao 266042 , P. R. China
| | - Haozheng Wang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , Jilin 130022 , China
- Key Laboratory of Olefin Catalysis and Polymerization/Key Laboratory of Rubber-Plastics (QUST) of Shandong Provincial , Qingdao University of Science and Technology , Qingdao 266042 , P. R. China
| | - Zehong Xiang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , Jilin 130022 , China
| | - Zhifang Ma
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , Jilin 130022 , China
| | - Jiruo Zhao
- Key Laboratory of Olefin Catalysis and Polymerization/Key Laboratory of Rubber-Plastics (QUST) of Shandong Provincial , Qingdao University of Science and Technology , Qingdao 266042 , P. R. China
| | - Ying Feng
- Key Laboratory of Olefin Catalysis and Polymerization/Key Laboratory of Rubber-Plastics (QUST) of Shandong Provincial , Qingdao University of Science and Technology , Qingdao 266042 , P. R. China
| | - Qiang Shi
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , Jilin 130022 , China
- University of Science and Technology of China , Hefei , Anhui 230026 , P. R. China
| | - Jinghua Yin
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry , Chinese Academy of Sciences , Changchun , Jilin 130022 , China
| |
Collapse
|
11
|
Konstandi M, Sotiropoulos I, Matsubara T, Malliou F, Katsogridaki A, Andriopoulou CE, Gonzalez FJ. Adrenoceptor-stimulated inflammatory response in stress-induced serum amyloid A synthesis. Psychopharmacology (Berl) 2019; 236:1687-1699. [PMID: 30612190 PMCID: PMC6643287 DOI: 10.1007/s00213-018-5149-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 12/11/2018] [Indexed: 12/20/2022]
Abstract
RATIONALE Stressful life events are suggested to contribute to the development of various pathologies, such as cardiovascular disorders, whose etiopathogenesis is highly associated with elevated levels of serum amyloid A (SAA) proteins. SAA synthesis in the liver is regulated by a complex network of cytokines acting independently or in concert with various hormones/stimulants including the stress-activated sympathetic nervous system. OBJECTIVE This study aims to investigate the underlying mechanisms that regulate the stress-induced hepatic synthesis of SAA, with particular focus on adrenoceptors (AR), major components of the sympathoadrenal response to stress. METHODS AND RESULTS We demonstrated that repeated stress elevates IL-1β, IL-6, and TNFα serum levels in mice, accompanied by increased synthesis and secretion of hepatic SAA1/2 and SAA3, an effect that was blocked by AR antagonists. Moreover, stimulation of α1- and β1/2-ARs mimics the stress effect on SAA1/2 regulation, whereas α2-AR stimulation exhibits a relatively weak impact on SAA. In support of the essential cytokine contribution in the AR-agonist induced SAA production is the fact that the anti-inflammatory drug, sodium salicylate, prevented the AR-stimulated hepatic SAA1/2 synthesis by reducing IL-1β levels, whereas IL-1β inhibition with Anakinra mimics this sodium salicylate preventive effect, thus indicating a crucial role for IL-1β. Interestingly, the AR-driven SAA3 synthesis was elevated by sodium salicylate in a TNFα-dependent way, supporting diverse and complex regulatory roles of cytokines in SAA production. In contrast to α1/α2-AR, the β1/2-AR-mediated SAA1/2 and SAA3 upregulation cannot be reversed by fenofibrate, a hypolipidemic drug with anti-inflammatory properties. CONCLUSION Taken together, these findings strongly support a critical role of the AR-stimulated inflammatory response in the hepatic SAA production under stressful conditions, highlighting distinct AR type-specific mechanisms that regulate the hepatic synthesis of SAA1/2 and SAA3.
Collapse
Affiliation(s)
- Maria Konstandi
- Faculty of Medicine, Department of Pharmacology, University of Ioannina, Ioannina, Greece. .,Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Ioannis Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), Medical School University of Minho, Braga, Portugal,ICVS/3B’s - PT Government Associate Laboratory, Braga, Portugal
| | - Tsutomu Matsubara
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA,Graduate School of Medicine, Department of Anatomy and Regenerative Biology, Osaka City University, Osaka, Japan
| | - Foteini Malliou
- Faculty of Medicine, Department of Pharmacology, University of Ioannina, Ioannina, Greece
| | - Alexandra Katsogridaki
- Faculty of Medicine, Department of Pharmacology, University of Ioannina, Ioannina, Greece
| | | | - Frank J. Gonzalez
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
12
|
LPS-Induced Systemic Inflammation Does Not Alter Atherosclerotic Plaque Area or Inflammation in APOE3∗LEIDEN Mice in the Early Phase Up to 15 Days. Shock 2018; 50:360-365. [DOI: 10.1097/shk.0000000000001026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
13
|
Li CY, Cui JY. Regulation of protein-coding gene and long noncoding RNA pairs in liver of conventional and germ-free mice following oral PBDE exposure. PLoS One 2018; 13:e0201387. [PMID: 30067809 PMCID: PMC6070246 DOI: 10.1371/journal.pone.0201387] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 07/14/2018] [Indexed: 02/07/2023] Open
Abstract
Gut microbiome communicates with the host liver to modify hepatic xenobiotic biotransformation and nutrient homeostasis. Polybrominated diphenyl ethers (PBDEs) are persistent environmental contaminants that are detected in fatty food, household dust, and human breast milk at worrisome levels. Recently, long noncoding RNAs (lncRNAs) have been recognized as novel biomarkers for toxicological responses and may regulate the transcriptional/translational output of protein-coding genes (PCGs). However, very little is known regarding to what extent the interactions between PBDEs and gut microbiome modulate hepatic lncRNAs and PCGs, and what critical signaling pathways are impacted at the transcriptomic scale. In this study, we performed RNA-Seq in livers of nine-week-old male conventional (CV) and germ-free (GF) mice orally exposed to the most prevalent PBDE congeners BDE-47 and BDE-99 (100 μmol/kg once daily for 4-days; vehicle: corn oil, 10 ml/kg), and unveiled key molecular pathways and PCG-lncRNA pairs targeted by PBDE-gut microbiome interactions. Lack of gut microbiome profoundly altered the PBDE-mediated transcriptomic response in liver, with the most prominent effect observed in BDE-99-exposed GF mice. The top pathways up-regulated by PBDEs were related to xenobiotic metabolism, whereas the top pathways down-regulated by PBDEs were in lipid metabolism and protein synthesis in both enterotypes. Genomic annotation of the differentially regulated lncRNAs revealed that majority of these lncRNAs overlapped with introns and 3'-UTRs of PCGs. Lack of gut microbiome profoundly increased the percentage of PBDE-regulated lncRNAs mapped to the 3'-UTRs of PCGs, suggesting the potential involvement of lncRNAs in increasing the translational efficiency of PCGs by preventing miRNA-3'-UTR binding, as a compensatory mechanism following toxic exposure to PBDEs. Pathway analysis of PCGs paired with lncRNAs revealed that in CV mice, BDE-47 regulated nucleic acid and retinol metabolism, as well as circadian rhythm; whereas BDE-99 regulated fatty acid metabolism. In GF mice, BDE-47 differentially regulated 19 lncRNA-PCG pairs that were associated with glutathione conjugation and transcriptional regulation. In contrast, BDE-99 up-regulated the xenobiotic-metabolizing Cyp3a genes, but down-regulated the fatty acid-metabolizing Cyp4 genes. Taken together, the present study reveals common and unique lncRNAs and PCG targets of PBDEs in mouse liver, and is among the first to show that lack of gut microbiome sensitizes the liver to toxic exposure of BDE-99 but not BDE-47. Therefore, lncRNAs may serve as specific biomarkers that differentiate various PBDE congeners as well as environmental chemical-mediated dysbiosis. Coordinate regulation of PCG-lncRNA pairs may serve as a more efficient molecular mechanism to combat against xenobiotic insult, and especially during dysbiosis-induced increase in the internal dose of toxicants.
Collapse
Affiliation(s)
- Cindy Yanfei Li
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
14
|
Noninvasive Imaging of Stored Red Blood Cell-Transfusion Aggravating Sepsis-Induced Liver Injury Associated with Increased Activation of M1-Polarized Kupffer Cells. Shock 2018; 48:459-466. [PMID: 28333715 PMCID: PMC5571877 DOI: 10.1097/shk.0000000000000867] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Supplemental Digital Content is available in the text Liver injury has a critical effect on the severity and outcome of sepsis. The impact of stored red blood cells (RBCs) on the pathogenesis of sepsis-associated hepatic injury is not well understood. Therefore, to investigate the effects of stored-RBC transfusion on sepsis-induced liver damage as well as the associated mechanism, we constructed a sepsis mouse model enabling noninvasive imaging of bacterial infection caused by Pseudomonas aeruginosa, a common gram-negative respiratory pathogen. We showed that transfusions with stored RBCs enhanced sepsis-induced liver injury in vivo, and liver injury exacerbated the severity of sepsis and decreased survival in P aeruginosa-infected mice. Stored-RBC transfusions enhanced the production of proinflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin 6 (IL-6), and IL-1β, which play important roles in sepsis-associated liver injury in P aeruginosa-infected mice. Further study showed that the enhanced inflammation observed was associated with increased activation of M1-polarized Kupffer cells, which produce many inflammatory cytokines, including TNF-α and IL-6. Moreover, the M1-polarized Kupffer cells and secreted proinflammatory cytokines exerted their effects on hepatocytes through enhanced Jun N-terminal kinase activation and inhibited nuclear factor-kappaB activation, demonstrating that transfusion with stored RBCs disrupted the balance between cell survival and cell death in the liver. Understanding the mechanisms whereby stored RBCs might contribute to these complications will likely be helpful in providing guidance toward making transfusions safer.
Collapse
|
15
|
Al-Shaebi EA, Dkhil MA, Al-Quraishy S. Indigofera oblongifolia regulates the hepatic gene expression profile induced by blood stage malaria. Microb Pathog 2018; 119:170-182. [PMID: 29630927 DOI: 10.1016/j.micpath.2018.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/03/2018] [Accepted: 04/05/2018] [Indexed: 11/25/2022]
Abstract
Malaria is still a major health problem worldwide. This study aimed to investigate the hepatoprotective role of Indigofera oblongifolia leaf extracts (ILE) against mice hepatic injury induced by Plasmodium chabaudi. Female C57BL/6 mice were treated with 100 mg/kg of ILE after infection with erythrocytes parasitized by P. chabaudi. On day 7 post-infection, the extract improved the histological alteration induced by the parasite. This was evidenced by the decreased histological index induced by ILE. Moreover, ILE was able to increase the hepatic antioxidant capacity and could significantly improve the decrease in erythrocyte count and hemoglobin content in mice blood plasma due to infection. ILE was also able to upregulate the expression of 24 genes related to metabolism and of 3 genes related to the immune response. Furthermore, the extract was able to downregulate the expression of 35 genes related to metabolism and of 82 genes related to immune response. Moreover, the microarray study showed that ILE regulated the change in gene expression induced by the parasite. Among these genes, we quantified the expression of cd209f, cyp7a1, Hsd3b5, Sult2a3, Lcn2, CcI8, Nos2, and saa3-mRNAs. These genes were regulated by ILE. Therefore, our results revealed the protective role of Indigofera oblongifolia against hepatic injury induced by blood stage malaria.
Collapse
Affiliation(s)
- Esam A Al-Shaebi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed A Dkhil
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia; Department of Zoology and Entomology, College of Science, Helwan University, Cairo, Egypt
| | - Saleh Al-Quraishy
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
16
|
Sugimura T, Jounai K, Ohshio K, Suzuki H, Kirisako T, Sugihara Y, Fujiwara D. Long-term administration of pDC-Stimulative Lactococcus lactis strain decelerates senescence and prolongs the lifespan of mice. Int Immunopharmacol 2018; 58:166-172. [PMID: 29605632 DOI: 10.1016/j.intimp.2018.03.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/01/2018] [Accepted: 03/23/2018] [Indexed: 10/17/2022]
Abstract
The decline in immune function caused by aging increases the risk of infectious diseases, tumorigeneses and chronic inflammation, resulting in accelerating senescence. We previously reported a lactic acid bacteria, Lactococcus lactis strain Plasma (synonym of Lactococcus lactis subsp. lactis JCM 5805, Lc-Plasma), that stimulates plasmacytoid dendritic cells (pDCs), which play a crucial role in phylaxis from viral infection. In this study, we investigated the anti-aging effects of long-term oral administration of Lc-Plasma in a senescence-accelerated mouse strain, SAMP6. Mice given Lc-Plasma showed a significant improvement in survival rate at 82 weeks and a decreased senescence score as compared with control mice throughout this study. Anatomic analysis at 82 weeks revealed that the frequency of altered hepatocellular foci was significantly lower, and the incidence of other pathological findings in the liver and lungs tended to be lower in Lc-Plasma mice than in control mice. Transcription level of the IL-1β gene in lungs also tended to be lower in Lc-Plasma mice. Furthermore, the thinning of skin and age-related decrease in muscle mass were also significantly suppressed in the Lc-Plasma group as compared with the control group. Consistent with these phenotypic features, pDCs activity was significantly higher in Lc-Plasma mice than in control mice. In conclusion, long-term administration of Lc-Plasma can decelerate senescence and prolong lifespan via maintenance of the immune system due to activation of pDCs.
Collapse
Affiliation(s)
- Tetsu Sugimura
- Central Laboratories for Key Technologies, Kirin Co. Ltd., Yokohama, Japan.
| | - Kenta Jounai
- Central Laboratories for Key Technologies, Kirin Co. Ltd., Yokohama, Japan; Technical Deveropment Center, Koiwai Dairy Products Co Ltd., Sayama, Japan
| | - Konomi Ohshio
- Central Laboratories for Key Technologies, Kirin Co. Ltd., Yokohama, Japan
| | - Hiroaki Suzuki
- Central Laboratories for Key Technologies, Kirin Co. Ltd., Yokohama, Japan
| | - Takayoshi Kirisako
- Central Laboratories for Key Technologies, Kirin Co. Ltd., Yokohama, Japan
| | - Yoshihiko Sugihara
- Central Laboratories for Key Technologies, Kirin Co. Ltd., Yokohama, Japan
| | - Daisuke Fujiwara
- Central Laboratories for Key Technologies, Kirin Co. Ltd., Yokohama, Japan
| |
Collapse
|
17
|
Tiwari A, Singh P, Jaitley P, Sharma S, Prakash A, Mandil R, Choudhury S, Gangwar NK, Garg SK. Eucalyptus robusta leaves methanolic extract suppresses inflammatory mediators by specifically targeting TLR4/TLR9, MPO, COX2, iNOS and inflammatory cytokines in experimentally-induced endometritis in rats. JOURNAL OF ETHNOPHARMACOLOGY 2018; 213:149-158. [PMID: 29104078 DOI: 10.1016/j.jep.2017.10.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/31/2017] [Accepted: 10/31/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bacterial endometritis is one of the major causes of reproductive disorders including infertility in farm animals. Antibiotics are generally used for treatment of such disorders but now a days residues of antibiotics are of great public health concern, therefore, phytoremediation is being considered as an alternative to use of antibiotics. AIM OF THE STUDY Present study was undertaken to investigate the efficacy of Eucalyptus robusta leaves methanolic extract against endometritis along with the possible mechanism of action especially targeting inflammatory biomarkers. MATERIALS AND METHODS Bacterial endometritis was produced using clinical isolates of E. coli and Staphyloccocus aureus from bovines (cows and buffaloes) endometritis cases. After seven days of inoculation of the mixed bacterial culture, endometritis was confirmed based on the presence of visible pus and edema, thinning of endometrial lining and presence of large number of polymorphonuclear cells and bacterial load in uterine flushing. Female Wistar rats were divided in to five groups namely control, sham-operated, endometritis, endometritis plus Eucalyptus leaves extract and endometritis plus cefixime. Serum specific inflammatory biomarkers (interleukin-1β, interleukin-10, tumor necrosis factor-α, intercellular adhesion molecule-1, serum amyloid A) and myleoperoxidase, toll like receptors-4 and -9, inducible nitric oxide synthase, nitric oxide, cyclooxygenase 1 and 2 were estimated in uterine tissues using ELISA kits. RESULTS Interleukin-10, serum amyloid A, myleoperoxidase, toll like receptors-4 and-9, cyclooxygenase-2, inducible nitric oxide synthase and nitric oxide were significantly increased while non significant increase in interleukin-1β, cycloxygenase-1 and intercellular adhesion molecule-1 were observed but level of tumor necrosis factor-α was found decreased in rats of endometritis group. Histopathological lesions in uterus showed efficient induction of endometritis by presence of inflammatory cells which are lessened effectively after treatment with Eucalyptus leaves extract. Eucalyptus robusta leaves extract produced curative and protective effect against endometritis and results were comparable to or even better than cefixime. CONCLUSIONS Eucalyptus robusta leaves extract possess promising antibacterial activity and efficacy against experimental endometritis and, therefore, holds promising potential for development of effective formulation for treatment of endometritis in animals.
Collapse
Affiliation(s)
- Aastha Tiwari
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pt. Deen Dayal Upadhyay Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India.
| | - Preeti Singh
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pt. Deen Dayal Upadhyay Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India.
| | - Pooja Jaitley
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pt. Deen Dayal Upadhyay Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India.
| | - Sushant Sharma
- Department of Livestock Products Technology, College of Veterinary Science and Animal Husbandry, U.P. Pt. Deen Dayal Upadhyay Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India.
| | - Atul Prakash
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pt. Deen Dayal Upadhyay Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India.
| | - Rajesh Mandil
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pt. Deen Dayal Upadhyay Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India.
| | - Soumen Choudhury
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pt. Deen Dayal Upadhyay Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India.
| | - Neeraj Kumar Gangwar
- Department of Veterinary Pathology, College of Veterinary Science and Animal Husbandry, U.P. Pt. Deen Dayal Upadhyay Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India.
| | - Satish K Garg
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pt. Deen Dayal Upadhyay Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India.
| |
Collapse
|
18
|
Zhu Q, Wu N, Liu G, Zhou Y, Liu S, Chen J, Liu J, Zuo Y, Liu Z, Chen W, Chen Y, Chen J, Lin M, Zhao Y, Yang Y, Wang S, Yang X, Ma Y, Wang J, Chen X, Zhang J, Shen J, Wu Z, Qiu G. Comparative analysis of serum proteome in congenital scoliosis patients with TBX6 haploinsufficiency - a first report pointing to lipid metabolism. J Cell Mol Med 2017; 22:533-545. [PMID: 28944995 PMCID: PMC5742745 DOI: 10.1111/jcmm.13341] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 06/24/2017] [Indexed: 12/17/2022] Open
Abstract
Congenital scoliosis (CS) is a three‐dimensional deformity of the spine affecting quality of life. We have demonstrated TBX6 haploinsufficiency is the most important contributor to CS. However, the pathophysiology at the protein level remains unclear. Therefore, this study was to explore the differential proteome in serum of CS patients with TBX6 haploinsufficiency. Sera from nine CS patients with TBX6 haploinsufficiency and nine age‐ and gender‐matched healthy controls were collected and analysed by isobaric tagged relative and absolute quantification (iTRAQ) labelling coupled with mass spectrometry (MS). In total, 277 proteins were detected and 20 proteins were designated as differentially expressed proteins, which were submitted to subsequent bioinformatics analysis. Gene Ontology classification analysis showed the biological process was primarily related to ‘cellular process’, molecular function ‘structural molecule activity’ and cellular component ‘extracellular region’. IPA analysis revealed ‘LXR/RXR activation’ was the top pathway, which is a crucial pathway in lipid metabolism. Hierarchical clustering analysis generated two clusters. In summary, this study is the first proteomic research to delineate the total and differential serum proteins in TBX6 haploinsufficiency‐caused CS. The proteins discovered in this experiment may serve as potential biomarkers for CS, and lipid metabolism might play important roles in the pathogenesis of CS.
Collapse
Affiliation(s)
- Qiankun Zhu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Nan Wu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Research Center of Orthopedics/Rare Disease, Chinese Academy of Medical Sciences, Beijing, China
| | - Gang Liu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Research Center of Orthopedics/Rare Disease, Chinese Academy of Medical Sciences, Beijing, China
| | - Yangzhong Zhou
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Tsinghua University Medical School, Beijing, China
| | - Sen Liu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Research Center of Orthopedics/Rare Disease, Chinese Academy of Medical Sciences, Beijing, China
| | - Jun Chen
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jiaqi Liu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yuzhi Zuo
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Research Center of Orthopedics/Rare Disease, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhenlei Liu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Weisheng Chen
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Yixin Chen
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jia Chen
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Mao Lin
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China
| | - Yanxue Zhao
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Yang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Shensgru Wang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xu Yang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yufen Ma
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jian Wang
- Department of Medical Genetics, Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoli Chen
- Department of Medical Genetics, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Jianguo Zhang
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jianxiong Shen
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhihong Wu
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Research Center of Orthopedics/Rare Disease, Chinese Academy of Medical Sciences, Beijing, China.,Department of Central Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Guixing Qiu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Beijing, China.,Research Center of Orthopedics/Rare Disease, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
19
|
Antioxidative activity of high-density lipoprotein (HDL): Mechanistic insights into potential clinical benefit. BBA CLINICAL 2017; 8:66-77. [PMID: 28936395 PMCID: PMC5597817 DOI: 10.1016/j.bbacli.2017.07.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 12/29/2022]
Abstract
Uptake of low-density lipoprotein (LDL) particles by macrophages represents a key step in the development of atherosclerotic plaques, leading to the foam cell formation. Chemical modification of LDL is however necessary to induce this process. Proatherogenic LDL modifications include aggregation, enzymatic digestion and oxidation. LDL oxidation by one-electron (free radicals) and two-electron oxidants dramatically increases LDL affinity to macrophage scavenger receptors, leading to rapid LDL uptake and fatty streak formation. Circulating high-density lipoprotein (HDL) particles, primarily small, dense, protein-rich HDL3, provide potent protection of LDL from oxidative damage by free radicals, resulting in the inhibition of the generation of pro-inflammatory oxidized lipids. HDL-mediated inactivation of lipid hydroperoxides involves their initial transfer from LDL to HDL and subsequent reduction to inactive hydroxides by redox-active Met residues of apolipoprotein A-I. Several HDL-associated enzymes are present at elevated concentrations in HDL3 relative to large, light HDL2 and can be involved in the inactivation of short-chain oxidized phospholipids. Therefore, HDL represents a multimolecular complex capable of acquiring and inactivating proatherogenic lipids. Antioxidative function of HDL can be impaired in several metabolic and inflammatory diseases. Structural and compositional anomalies in the HDL proteome and lipidome underlie such functional deficiency. Concomitant normalization of the metabolism, circulating levels, composition and biological activities of HDL particles, primarily those of small, dense HDL3, can constitute future therapeutic target.
Collapse
|
20
|
Erickson MA, Jude J, Zhao H, Rhea EM, Salameh TS, Jester W, Pu S, Harrowitz J, Nguyen N, Banks WA, Panettieri RA, Jordan-Sciutto KL. Serum amyloid A: an ozone-induced circulating factor with potentially important functions in the lung-brain axis. FASEB J 2017; 31:3950-3965. [PMID: 28533327 DOI: 10.1096/fj.201600857rrr] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 05/01/2017] [Indexed: 01/24/2023]
Abstract
Accumulating evidence suggests that O3 exposure may contribute to CNS dysfunction. Here, we posit that inflammatory and acute-phase proteins in the circulation increase after O3 exposure and systemically convey signals of O3 exposure to the CNS. To model acute O3 exposure, female Balb/c mice were exposed to 3 ppm O3 or forced air for 2 h and were studied after 6 or 24 h. Of 23 cytokines and chemokines, only KC/CXCL1 was increased in blood 6 h after O3 exposure. The acute-phase protein serum amyloid A (A-SAA) was significantly increased by 24 h, whereas C-reactive protein was unchanged. A-SAA in blood correlated with total leukocytes, macrophages, and neutrophils in bronchoalveolar lavage from O3-exposed mice. A-SAA mRNA and protein were increased in the liver. We found that both isoforms of A-SAA completely crossed the intact blood-brain barrier, although the rate of SAA2.1 influx was approximately 5 times faster than that of SAA1.1. Finally, A-SAA protein, but not mRNA, was increased in the CNS 24 h post-O3 exposure. Our findings suggest that A-SAA is functionally linked to pulmonary inflammation in our O3 exposure model and that A-SAA could be an important systemic signal of O3 exposure to the CNS.-Erickson, M. A., Jude, J., Zhao, H., Rhea, E. M., Salameh, T. S., Jester, W., Pu, S., Harrowitz, J., Nguyen, N., Banks, W. A., Panettieri, R. A., Jr., Jordan-Sciutto, K. L. Serum amyloid A: an ozone-induced circulating factor with potentially important functions in the lung-brain axis.
Collapse
Affiliation(s)
- Michelle A Erickson
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; .,Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Joseph Jude
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey, USA.,Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hengjiang Zhao
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth M Rhea
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Therese S Salameh
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - William Jester
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey, USA.,Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shelley Pu
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jenna Harrowitz
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ngan Nguyen
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William A Banks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Reynold A Panettieri
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.,Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kelly L Jordan-Sciutto
- Department of Pathology, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Türer ÇC, Ballı U, Güven B. Fetuin-A, serum amyloid A and tumor necrosis factor alpha levels in periodontal health and disease. Oral Dis 2017; 23:379-386. [DOI: 10.1111/odi.12625] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/24/2016] [Accepted: 12/13/2016] [Indexed: 12/19/2022]
Affiliation(s)
- ÇC Türer
- Department of Periodontology; Faculty of Dentistry; Bulent Ecevit University; Zonguldak Turkey
| | - U Ballı
- Department of Periodontology; Faculty of Dentistry; Bulent Ecevit University; Zonguldak Turkey
| | - B Güven
- Department of Biochemistry; Faculty of Medicine; Bulent Ecevit University; Zonguldak Turkey
| |
Collapse
|
22
|
Izumi H, Ishimoto T, Yamamoto H, Mori H. Application of hairless mouse strain to bioluminescence imaging of Arc expression in mouse brain. BMC Neurosci 2017; 18:18. [PMID: 28114886 PMCID: PMC5260114 DOI: 10.1186/s12868-017-0335-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 01/12/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Bioluminescence imaging (BLI) is a powerful technique for monitoring the temporal and spatial dynamics of gene expression in the mouse brain. However, the black fur, skin pigmentation and hair regrowth after depilation of mouse interfere with BLI during developmental and daily examination. The aim of this study was to extend the application of Arc-Luc transgenic (Tg) mice to the BLI of neuronal activity in the mouse brain by introducing the hairless (HL) gene and to examine Arc-Luc expression at various developmental stages without interference from black fur, skin pigmentation, and hair regrowth. RESULTS The Arc-Luc Tg HL mice were established by crossing the Tg C57BL/6 mouse strain with the HL mouse strain. Under physiological and pathological conditions, BLI was performed to detect the signal intensity changes at various developmental stages and at an interval of <7 days. The established Arc-Luc Tg HL mice exhibited clear and stable photon signals from the brain without interference during development. After surgical monocular deprivation during visual-critical period, large signal intensity changes in bioluminescence were observed in the mouse visual cortex. Exposure of mice to a novel object changed the photon distribution in the caudal and rostral cerebral areas. The temporal pattern of kainic-acid-induced Arc-Luc expression showed biphasic changes in signal intensity over 24 h. CONCLUSIONS This study showed the advantages of using the mutant HL gene in BLI of Arc expression in the mouse brain at various developmental stages. Thus, the use of the Arc-Luc Tg HL mice enabled the tracking of neuronal-activity-dependent processes over a wide range from a focal area to the entire brain area with various time windows.
Collapse
Affiliation(s)
- Hironori Izumi
- Department of Molecular Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Tetsuya Ishimoto
- Department of Molecular Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Hiroshi Yamamoto
- Division of Animal Resources and Development, Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
23
|
Gandhi D, Naoghare PK, Bafana A, Kannan K, Sivanesan S. Fluoride-Induced Oxidative and Inflammatory Stress in Osteosarcoma Cells: Does It Affect Bone Development Pathway? Biol Trace Elem Res 2017; 175:103-111. [PMID: 27234253 DOI: 10.1007/s12011-016-0756-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 05/20/2016] [Indexed: 12/30/2022]
Abstract
Oxidative stress is reported to negatively affect osteoblast cells. Present study reports oxidative and inflammatory signatures in fluoride-exposed human osteosarcoma (HOS) cells, and their possible association with the genes involved in osteoblastic differentiation and bone development pathways. HOS cells were challenged with sublethal concentration (8 mg/L) of sodium fluoride for 30 days and analyzed for transcriptomic expression. In total, 2632 transcripts associated with several biological processes were found to be differentially expressed. Specifically, genes involved in oxidative stress, inflammation, osteoblastic differentiation, and bone development pathways were found to be significantly altered. Variation in expression of key genes involved in the abovementioned pathways was validated through qPCR. Expression of serum amyloid A1 protein, a key regulator of stress and inflammatory pathways, was validated through western blot analysis. This study provides evidence that chronic oxidative and inflammatory stress may be associated with the fluoride-induced impediment in osteoblast differentiation and bone development.
Collapse
Affiliation(s)
- Deepa Gandhi
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Pravin K Naoghare
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Amit Bafana
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Krishnamurthi Kannan
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Saravanadevi Sivanesan
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India.
| |
Collapse
|
24
|
Fuijkschot WW, Morrison MC, van der Linden R, Krijnen PAJ, Zethof IPA, Theyse LFH, Kleemann R, Niessen HWM, Smulders YM. Orthopedic surgery increases atherosclerotic lesions and necrotic core area in ApoE-/- mice. Atherosclerosis 2016; 255:164-170. [PMID: 27825629 DOI: 10.1016/j.atherosclerosis.2016.07.909] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 07/19/2016] [Accepted: 07/20/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIMS Observational studies show a peak incidence of cardiovascular events after major surgery. For example, the risk of myocardial infarction increases 25-fold early after hip replacement. The acuteness of this increased risk suggests abrupt enhancement in plaque vulnerability, which may be related to intra-plaque inflammation, thinner fibrous cap and/or necrotic core expansion. We hypothesized that acute systemic inflammation following major orthopedic surgery induces such changes. METHODS ApoE-/- mice were fed a western diet for 10 weeks. Thereafter, half the mice underwent mid-shaft femur osteotomy followed by realignment with an intramedullary K-wire, to mimic major orthopedic surgery. Mice were sacrificed 5 or 15 days post-surgery (n = 22) or post-saline injection (n = 13). Serum amyloid A (SAA) was measured as a marker of systemic inflammation. Paraffin embedded slides of the aortic root were stained to measure total plaque area and to quantify fibrosis, calcification, necrotic core, and inflammatory cells. RESULTS Surgery mice showed a pronounced elevation of serum amyloid A (SAA) and developed increased plaque and necrotic core area already at 5 days, which reached significance at 15 days (p = 0.019; p = 0.004 for plaque and necrotic core, respectively). Macrophage and lymphocyte density significantly decreased in the surgery group compared to the control group at 15 days (p = 0.037; p = 0.024, respectively). The density of neutrophils and mast cells remained unchanged. CONCLUSIONS Major orthopedic surgery in ApoE-/- mice triggers a systemic inflammatory response. Atherosclerotic plaque area is enlarged after surgery mainly due to an increase of the necrotic core. The role of intra-plaque inflammation in this response to surgical injury remains to be fully elucidated.
Collapse
Affiliation(s)
- Wessel W Fuijkschot
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands; Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands.
| | - Martine C Morrison
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | | | - Paul A J Krijnen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands; Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands
| | - Ilse P A Zethof
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | - Lars F H Theyse
- Department Clinical Sciences and Services, Royal Veterinary College London, University of London, United Kingdom
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands; Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans W M Niessen
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands; Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; Department of Cardiac Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Yvo M Smulders
- Institute for Cardiovascular Research (ICaR-VU), VU University Medical Center, Amsterdam, The Netherlands; Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Zhang Y, Zhou Q, Yan S, Zhang N, Zhao M, Ma C, He C, Fu Q, Wu T, Wang X, Zhan L. Non-Invasive Imaging Serum Amyloid A Activation through the NF-κB Signal Pathway upon Gold Nanostructure Exposure. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:3270-3282. [PMID: 27167493 DOI: 10.1002/smll.201600019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/01/2016] [Indexed: 06/05/2023]
Abstract
With the objective of investigating the acute activation of inflammatory cascades upon exposure to gold nanoparticles (GNPs) as well as detailing the mechanisms, a reporter mouse model that allows for non-invasive and longitudinal imaging of hepatic acute-phase serum amyloid A (SAA) activation is constructed. The model is able to visualize SAA activation at the transcriptional stage, with higher sensitivity than serum protein detection by ELISA. GNPs of various sizes (10-80 nm) and geometries are assessed using the reporter mice with results demonstrating that 50 nm nanospheres (GNS50) possess the highest capacity to induce hepatic SAA activation. Detailed analysis uncovers that resident macrophages in the liver are the main origins of these cytokines and that the exposure to GNS50 significantly induces the M1 macrophage phenotype. Moreover, those M1-polarized macrophages, together with the subsequently secreted pro-inflammatory cytokines, exert effects on hepatocytes and then initiate SAA transcription through the NF-κB signal pathway. The results detail the sequential reactions to GNPs among macrophages, inflammatory mediators, and SAA-synthesizing hepatocytes, which shed light on the acute effects of GNPs on the body. In addition, the established in situ and highly sensitive SAA detection system is expected to have vast applications in evaluating NP-induced acute inflammatory reactions.
Collapse
Affiliation(s)
- Yulong Zhang
- Beijing Institute of Transfusion Medicine, Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P. R. China
| | - Qianqian Zhou
- Beijing Institute of Transfusion Medicine, Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P. R. China
| | - Shaoduo Yan
- Beijing Institute of Transfusion Medicine, Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P. R. China
| | - Ning Zhang
- WuXi AppTec, Shanghai, 200131, P. R. China
| | - Man Zhao
- Beijing Institute of Transfusion Medicine, Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P. R. China
| | - Cong Ma
- Beijing Institute of Transfusion Medicine, Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P. R. China
| | - Chulin He
- Beijing Institute of Transfusion Medicine, Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P. R. China
| | - Qiuxia Fu
- Beijing Institute of Transfusion Medicine, Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P. R. China
| | - Tao Wu
- Beijing Institute of Transfusion Medicine, Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P. R. China
| | - Xiaohui Wang
- Beijing Institute of Transfusion Medicine, Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P. R. China
- Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), Nankai University, Tianjin, 300071, China
| | - Linsheng Zhan
- Beijing Institute of Transfusion Medicine, Beijing Key Laboratory of Blood Safety and Supply Technologies, Beijing, 100850, P. R. China
| |
Collapse
|
26
|
Heyman-Lindén L, Kotowska D, Sand E, Bjursell M, Plaza M, Turner C, Holm C, Fåk F, Berger K. Lingonberries alter the gut microbiota and prevent low-grade inflammation in high-fat diet fed mice. Food Nutr Res 2016; 60:29993. [PMID: 27125264 PMCID: PMC4850145 DOI: 10.3402/fnr.v60.29993] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 02/16/2016] [Accepted: 02/17/2016] [Indexed: 12/15/2022] Open
Abstract
Background The gut microbiota plays an important role in the development of obesity and obesity-associated impairments such as low-grade inflammation. Lingonberries have been shown to prevent diet-induced obesity and low-grade inflammation. However, it is not known whether the effect of lingonberry supplementation is related to modifications of the gut microbiota. The aim of the present study was to describe whether consumption of different batches of lingonberries alters the composition of the gut microbiota, which could be relevant for the protective effect against high fat (HF)-induced metabolic alterations. Methods Three groups of C57BL/6J mice were fed HF diet with or without a supplement of 20% lingonberries from two different batches (Lingon1 and Lingon2) during 11 weeks. The composition and functionality of the cecal microbiota were assessed by 16S rRNA sequencing and PICRUSt. In addition, parameters related to obesity, insulin sensitivity, hepatic steatosis, inflammation and gut barrier function were examined. Results HF-induced obesity was only prevented by the Lingon1 diet, whereas both batches of lingonberries reduced plasma levels of markers of inflammation and endotoxemia (SAA and LBP) as well as modified the composition and functionality of the gut microbiota, compared to the HF control group. The relative abundance of Akkermansia and Faecalibacterium, genera associated with healthy gut mucosa and anti-inflammation, was found to increase in response to lingonberry intake. Conclusions Our results show that supplementation with lingonberries to an HF diet prevents low-grade inflammation and is associated with significant changes of the microbiota composition. Notably, the anti-inflammatory properties of lingonberries seem to be independent of effects on body weight gain.
Collapse
Affiliation(s)
| | - Dorota Kotowska
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Elin Sand
- ImaGene-IT, Medicon Village, Lund, Sweden
| | | | - Merichel Plaza
- Department of Chemistry, Center for Analysis and Synthesis, Lund University, Lund, Sweden
| | - Charlotta Turner
- Department of Chemistry, Center for Analysis and Synthesis, Lund University, Lund, Sweden
| | - Cecilia Holm
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Frida Fåk
- Food for Health Science Centre, Lund University, Medicon Village, Lund, Sweden
| | - Karin Berger
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
27
|
Barber AE, Fleming BA, Mulvey MA. Similarly Lethal Strains of Extraintestinal Pathogenic Escherichia coli Trigger Markedly Diverse Host Responses in a Zebrafish Model of Sepsis. mSphere 2016; 1:e00062-16. [PMID: 27303721 PMCID: PMC4894679 DOI: 10.1128/msphere.00062-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 03/29/2016] [Indexed: 02/06/2023] Open
Abstract
In individuals with sepsis, the infecting microbes are commonly viewed as generic inducers of inflammation while the host background is considered the primary variable affecting disease progression and outcome. To study the effects of bacterial strain differences on the maladaptive immune responses that are induced during sepsis, we employed a novel zebrafish embryo infection model using extraintestinal pathogenic Escherichia coli (ExPEC) isolates. These genetically diverse pathogens are a leading cause of sepsis and are becoming increasingly dangerous because of the rise of multidrug-resistant strains. Zebrafish infected with ExPEC isolates exhibit many of the pathophysiological features seen in septic human patients, including dysregulated inflammatory responses (cytokine storms), tachycardia, endothelial leakage, and progressive edema. However, only a limited subset of ExPEC isolates can trigger a sepsis-like state and death of the host when introduced into the bloodstream. Mirroring the situation in human patients, antibiotic therapy reduced ExPEC titers and improved host survival rates but was only effective within limited time frames that varied, depending on the infecting pathogen. Intriguingly, we find that phylogenetically distant but similarly lethal ExPEC isolates can stimulate markedly different host transcriptional responses, including disparate levels of inflammatory mediators. These differences correlate with the amounts of bacterial flagellin expression during infection, as well as differential activation of Toll-like receptor 5 by discrete flagellar serotypes. Altogether, this work establishes zebrafish as a relevant model of key aspects of human sepsis and highlights the ability of genetically distinct ExPEC isolates to induce divergent host responses independently of baseline host attributes. IMPORTANCE Sepsis is a life-threatening systemic inflammatory condition that is initiated by the presence of microorganisms in the bloodstream. In the United States, sepsis due to ExPEC and other pathogens kills well over a quarter of a million people each year and is associated with tremendous health care costs. A high degree of heterogeneity in the signs and symptomology of sepsis makes this disease notoriously difficult to effectively diagnose and manage. Here, using a zebrafish model of sepsis, we find that similarly lethal but genetically distinct ExPEC isolates can elicit notably disparate host responses. These variances are in part due to differences in the levels and types of flagellin that are expressed by the infecting ExPEC strains. A better understanding of the variable impact that bacterial factors like flagellin have on host responses during sepsis could lead to improved diagnostic and therapeutic approaches to these often deadly infections. Podcast: A podcast concerning this article is available.
Collapse
Affiliation(s)
- Amelia E Barber
- Division of Microbiology and Immunology, Pathology Department, University of Utah, Salt Lake City, Utah, USA
| | - Brittany A Fleming
- Division of Microbiology and Immunology, Pathology Department, University of Utah, Salt Lake City, Utah, USA
| | - Matthew A Mulvey
- Division of Microbiology and Immunology, Pathology Department, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
28
|
Dkhil MA, Lubbad MY, Al-Shaebi EM, Delic D, Al-Quraishy S. The antiplasmodial and spleen protective role of crude Indigofera oblongifolia leaf extract traditionally used in the treatment of malaria in Saudi Arabia. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:6235-46. [PMID: 26648699 PMCID: PMC4664501 DOI: 10.2147/dddt.s94673] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Malaria is one of the most serious natural hazards faced by human society. Although plant leaves of Indigofera oblongifolia have been used for the treatment of malaria in Saudi Arabian society, there is no laboratory-based evidence for the effectiveness and safety of the plant. This study therefore was designed to investigate the antimalarial and spleen protective activity of I. oblongifolia leaf extract (IOLE) in mice. Three doses (100, 200 and 300 mg/kg) of IOLE were used to treat mice infected with Plasmodium chabaudi-parasitized erythrocytes. The suppressive effect produced by the 100 mg/kg dose on parasitemia was highly significant compared to the infected nontreated group. This dose was also able to repair the change in the thickness of the mice spleen and significantly lower the number of apoptotic cells in the spleen. Moreover, I. oblongifolia also altered gene expression in the infected spleen. On day 7 postinfection, the mRNA expression of six genes – with immune response functions – was upregulated by more than twofold, while that of 24 other genes was downregulated. Among the differentially up- and downregulated genes under the effect of IOLE, we quantified the expression of Ccl8, Saa3, Cd209a, and Cd209b mRNAs. The expression data, determined by microarrays, were largely consistent with the expression analyses we performed with several arbitrarily selected genes using quantitative polymerase chain reaction (PCR). Based on our results, I. oblongifolia exhibits antimalarial activity and could protect the spleen from P. chabaudi-induced injury.
Collapse
Affiliation(s)
- Mohamed A Dkhil
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia ; Department of Zoology and Entomology, Faculty of Science, Helwan University, Helwan, Egypt
| | - Mahmoud Y Lubbad
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia ; General Directorate of Environmental and Occupational Health, Public Health Agency, Ministry of Health, Riyadh, Saudi Arabia
| | - Esam M Al-Shaebi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Denis Delic
- Boehringer-Ingelheim Pharma, Biberach, Germany
| | - Saleh Al-Quraishy
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
29
|
Pfensig C, Dominik A, Borufka L, Hinz M, Stange J, Eggert M. A New Application for Albumin Dialysis in Extracorporeal Organ Support: Characterization of a Putative Interaction Between Human Albumin and Proinflammatory Cytokines IL-6 and TNFα. Artif Organs 2015; 40:397-402. [PMID: 26365493 DOI: 10.1111/aor.12557] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Albumin dialysis in extracorporeal organ support is often performed in the treatment of liver failure as it facilitates the removal of toxic components from the blood. Here, we describe a possible effect of albumin dialysis on proinflammatory cytokine levels in vitro. Initially, albumin samples were incubated with different amounts of cytokines and analyzed by enzyme-linked immunosorbent assay (ELISA). Analysis of interleukin 6 (IL-6) and tumor necrosis factor alpha (TNFα) levels indicated that increased concentrations of albumin reduce the measureable amount of the respective cytokines. This led to the hypothesis that the used proinflammatory cytokines may interact with albumin. Size exclusion chromatography of albumin spiked with cytokines was carried out using high-performance liquid chromatography analysis. The corresponding fractions were evaluated by immunoblotting. We detected albumin and cytokines in the same fractions indicating an interaction of the small-sized cytokines IL-6 and TNFα with the larger-sized albumin. Finally, a two-compartment albumin dialysis in vitro model was used to analyze the effect of albumin on proinflammatory cytokines in the recirculation circuit during 6-h treatment. These in vitro albumin dialysis experiments indicated a significant decrease of IL-6, but not of TNFα, when albumin was added to the dialysate solution. Taken together, we were able to show a putative in vitro interaction of human albumin with the proinflammatory cytokine IL-6, but with less evidence for TNFα, and demonstrated an additional application for albumin dialysis in liver support therapy where IL-6 removal might be indicated.
Collapse
Affiliation(s)
- Claudia Pfensig
- Center for Extracorporeal Organ Support (CEOS), Department of Internal Medicine, University of Rostock, Rostock, Germany
| | - Adrian Dominik
- Center for Extracorporeal Organ Support (CEOS), Department of Internal Medicine, University of Rostock, Rostock, Germany
| | - Luise Borufka
- Center for Extracorporeal Organ Support (CEOS), Department of Internal Medicine, University of Rostock, Rostock, Germany
| | - Michael Hinz
- Division of Nephrology, Department of Medicine, Rostock University Medical Center, Rostock, Germany
| | - Jan Stange
- Division of Nephrology, Department of Medicine, Rostock University Medical Center, Rostock, Germany
| | - Martin Eggert
- Center for Extracorporeal Organ Support (CEOS), Department of Internal Medicine, University of Rostock, Rostock, Germany
| |
Collapse
|
30
|
Berry intake changes hepatic gene expression and DNA methylation patterns associated with high-fat diet. J Nutr Biochem 2015; 27:79-95. [PMID: 26423886 DOI: 10.1016/j.jnutbio.2015.08.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 08/17/2015] [Accepted: 08/18/2015] [Indexed: 12/13/2022]
Abstract
The liver is a critical organ for regulation of energy homeostasis and fatty liver disease is closely associated with obesity and insulin resistance. We have previously found that lingonberries, blackcurrants and bilberries prevent, whereas açai berries exacerbate, the development of hepatic steatosis and obesity in the high-fat (HF)-fed C57BL/6J mouse model. In this follow-up study, we investigated the mechanisms behind these effects. Genome-wide hepatic gene expression profiling indicates that the protective effects of lingonberries and bilberries are accounted for by several-fold downregulation of genes involved in acute-phase and inflammatory pathways (e.g. Saa1, Cxcl1, Lcn2). In contrast, açai-fed mice exhibit marked upregulation of genes associated with steatosis (e.g. Cfd, Cidea, Crat) and lipid and cholesterol biosynthesis, which is in line with the exacerbation of HF-induced hepatic steatosis in these mice. In silico transcription factor analysis together with immunoblot analysis identified NF-κB, STAT3 and mTOR as upstream regulators involved in mediating the observed transcriptional effects. To gain further insight into mechanisms involved in the gene expression changes, the HELP-tagging assay was used to identify differentially methylated CpG sites. Compared to the HF control group, lingonberries induced genome-wide hypermethylation and specific hypermethylation of Ncor2, encoding the corepressor NCoR/SMRT implicated in the regulation of pathways of metabolic homeostasis and inflammation. We conclude that the beneficial metabolic effects of lingonberries and bilberries are associated with downregulation of inflammatory pathways, whereas for blackcurrants, exerting similar metabolic effects, different mechanisms of action appear to dominate. NF-κB, STAT3 and mTOR are potential targets of the health-promoting effects of berries.
Collapse
|
31
|
Orabi AI, Sah S, Javed TA, Lemon KL, Good ML, Guo P, Xiao X, Prasadan K, Gittes GK, Jin S, Husain SZ. Dynamic imaging of pancreatic nuclear factor κB (NF-κB) activation in live mice using adeno-associated virus (AAV) infusion and bioluminescence. J Biol Chem 2015; 290:11309-20. [PMID: 25802340 PMCID: PMC4416837 DOI: 10.1074/jbc.m115.647933] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Indexed: 12/19/2022] Open
Abstract
Nuclear factor κB (NF-κB) is an important signaling molecule that plays a critical role in the development of acute pancreatitis. Current methods for examining NF-κB activation involve infection of an adenoviral NF-κB-luciferase reporter into cell lines or electrophoretic mobility shift assay of lysate. The use of adeno-associated viruses (AAVs) has proven to be an effective method of transfecting whole organs in live animals. We examined whether intrapancreatic duct infusion of AAV containing an NF-κB-luciferase reporter (AAV-NF-κB-luciferase) can reliably measure pancreatic NF-κB activation. We confirmed the infectivity of the AAV-NF-κB-luciferase reporter in HEK293 cells using a traditional luciferase readout. Mice were infused with AAV-NF-κB-luciferase 5 weeks before induction of pancreatitis (caerulein, 50 μg/kg). Unlike transgenic mice that globally express NF-κB-luciferase, AAV-infused mice showed a 15-fold increase in pancreas-specific NF-κB bioluminescence following 12 h of caerulein compared with baseline luminescence (p < 0.05). The specificity of the NF-κB-luciferase signal to the pancreas was confirmed by isolating the pancreas and adjacent organs and observing a predominant bioluminescent signal in the pancreas compared with liver, spleen, and stomach. A complementary mouse model of post-ERCP-pancreatitis also induced pancreatic NF-κB signals. Taken together these data provide the first demonstration that NF-κB activation can be examined in a live, dynamic fashion during pancreatic inflammation. We believe this technique offers a valuable tool to study real-time activation of NF-κB in vivo.
Collapse
Affiliation(s)
| | - Swati Sah
- From the Department of Pediatrics and
| | | | | | | | - Ping Guo
- Surgery, Children's Hospital of Pittsburgh of UPMC and the University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Xiangwei Xiao
- Surgery, Children's Hospital of Pittsburgh of UPMC and the University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Krishna Prasadan
- Surgery, Children's Hospital of Pittsburgh of UPMC and the University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - George K Gittes
- Surgery, Children's Hospital of Pittsburgh of UPMC and the University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | | | | |
Collapse
|
32
|
Chou AH, Lee CM, Chen CY, Liou JT, Liu FC, Chen YL, Day YJ. Hippocampal transcriptional dysregulation after renal ischemia and reperfusion. Brain Res 2014; 1582:197-210. [DOI: 10.1016/j.brainres.2014.07.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/12/2014] [Accepted: 07/17/2014] [Indexed: 11/26/2022]
|
33
|
Derebe MG, Zlatkov CM, Gattu S, Ruhn KA, Vaishnava S, Diehl GE, MacMillan JB, Williams NS, Hooper LV. Serum amyloid A is a retinol binding protein that transports retinol during bacterial infection. eLife 2014; 3:e03206. [PMID: 25073702 PMCID: PMC4129439 DOI: 10.7554/elife.03206] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Retinol plays a vital role in the immune response to infection, yet proteins that mediate retinol transport during infection have not been identified. Serum amyloid A (SAA) proteins are strongly induced in the liver by systemic infection and in the intestine by bacterial colonization, but their exact functions remain unclear. Here we show that mouse and human SAAs are retinol binding proteins. Mouse and human SAAs bound retinol with nanomolar affinity, were associated with retinol in vivo, and limited the bacterial burden in tissues after acute infection. We determined the crystal structure of mouse SAA3 at a resolution of 2 Å, finding that it forms a tetramer with a hydrophobic binding pocket that can accommodate retinol. Our results thus identify SAAs as a family of microbe-inducible retinol binding proteins, reveal a unique protein architecture involved in retinol binding, and suggest how retinol is circulated during infection. DOI:http://dx.doi.org/10.7554/eLife.03206.001 Vitamins are nutrients that organisms require in order to survive and grow. If an organism is unable to synthesize a vitamin in sufficient quantities, it is essential that it obtain the vitamin through its diet instead. Vitamin A is found in foods such as eggs, animal liver and carrots, and a diet that is lacking in this vitamin can cause blindness and an increased risk of microbial infections. Vitamin A is not a single compound, but rather a collection of compounds with similar molecular structures. One of these is retinol, which plays a vital role in the body's response to microbial infection. Retinol must bind to specific proteins to be able to move through the bloodstream and be transported around the body. Serum retinol binding protein transports ingested retinol from the intestine to the liver and other tissues. However, during microbial infection—when retinol transport is particularly important—the amount of this protein dramatically decreases; as such it is unclear how retinol is transported when the body is under attack from pathogens. It had been suggested that Serum Amyloid A (SAA) proteins, a family of proteins made by some liver and intestinal cells, could be involved in the response to infection, because these proteins' levels increase during infection. However, their exact functions were unknown. Derebe, Zlatkov et al. found that mice fed a diet poor in vitamin A produced fewer SAA proteins in their liver and intestinal cells. However, treating the cells with retinol or the molecule it is broken down into—called retinoic acid—caused more SAAs to be made. Derebe, Zlatkov et al. also discovered that SAAs are associated with retinol in blood samples taken from mice infected with salmonella; and that both mouse and human SAAs bind tightly to retinol. Combined, this evidence suggests that SAAs are the retinol binding proteins that transport retinol during infections. Derebe, Zlatkov et al. went on to solve the crystal structure of a mouse SAA protein, and showed that four SAA molecules bind together to form a ‘pocket’ that can hold a retinol molecule. Future work will focus on understanding exactly how the transport of retinol by SAAs affects the development of immunity to infections. DOI:http://dx.doi.org/10.7554/eLife.03206.002
Collapse
Affiliation(s)
- Mehabaw G Derebe
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Clare M Zlatkov
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Sureka Gattu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Kelly A Ruhn
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Shipra Vaishnava
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Gretchen E Diehl
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, United States
| | - John B MacMillan
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, United States
| | - Lora V Hooper
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, United States Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
34
|
Denervation protects limbs from inflammatory arthritis via an impact on the microvasculature. Proc Natl Acad Sci U S A 2014; 111:11419-24. [PMID: 25049388 DOI: 10.1073/pnas.1410854111] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Two-way communication between the mammalian nervous and immune systems is increasingly recognized and appreciated. An intriguing example of such crosstalk comes from clinical observations dating from the 1930s: Patients who suffer a stroke and then develop rheumatoid arthritis atypically present with arthritis on only one side, the one not afflicted with paralysis. Here we successfully modeled hemiplegia-induced protection from arthritis using the K/BxN serum-transfer system, focused on the effector phase of inflammatory arthritis. Experiments entailing pharmacological inhibitors, genetically deficient mouse strains, and global transcriptome analyses failed to associate the protective effect with a single nerve quality (i.e., with the sympathetic, parasympathetic, or sensory nerves). Instead, there was clear evidence that denervation had a long-term effect on the limb microvasculature: The rapid and joint-localized vascular leak that typically accompanies and promotes serum-transferred arthritis was compromised in denervated limbs. This defect was reflected in the transcriptome of endothelial cells, the expression of several genes impacting vascular leakage or transendothelial cell transmigration being altered in denervated limbs. These findings highlight a previously unappreciated pathway to dissect and eventually target in inflammatory arthritis.
Collapse
|
35
|
Choi S, Lim JY, Kim Y, Song MJ, Jung WW, Seo JB, Lee J, Sul D. Plasma proteomic analysis of patients infected with H1N1 influenza virus. Proteomics 2014; 14:1933-42. [PMID: 24888898 DOI: 10.1002/pmic.201400030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 05/05/2014] [Accepted: 05/23/2014] [Indexed: 11/10/2022]
Abstract
This study profiled the plasma proteins of patients infected by the 2011 H1N1 influenza virus. Differential protein expression was identified in plasma obtained from noninfected control subjects (n = 15) and H1N1-infected subjects (n = 15). Plasma proteins were separated by a 2DE large gel system and identified by nano-ultra performance LC-MS. Western blot assays were performed to validate proteins. Eight plasma proteins were upregulated and six proteins were downregulated among 3316 plasma proteins in the H1N1-infected group as compared with the control group. Of 14 up- and downregulated proteins, nine plasma proteins were validated by Western blot analysis. Putative protein FAM 157A, leucine-rich alpha 2 glycoprotein, serum amyloid A protein, and dual oxidase 1 showed significant differential expression. The identified plasma proteins could be potential candidates for biomarkers of H1N1 influenza viral infection. Further studies are needed to develop these proteins as diagnostic biomarkers.
Collapse
Affiliation(s)
- Seonyoung Choi
- Graduate School of Medicine, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
This review highlights the protective roles of bilirubin against the atherosclerotic process. Bilirubin belongs to the superfamily of tetrapyrrolic compounds formed during heme catabolism. Although for decades bilirubin was considered to be a harmful waste product, recent epidemiologic studies have shown that serum bilirubin levels have consistently been inversely associated with cardiovascular disease (CVD), as well as cardiovascular risk factors such as metabolic syndrome and diabetes. These clinical studies are supported by in vitro and in vivo experimental data and have demonstrated that bilirubin not only has an ability to scavenge overproduced reactive oxygen species and inhibit vascular smooth muscle cell proliferation but, additionally, has anti-inflammatory effects. In this review, we will discuss the inverse association of serum bilirubin and CVD and cardiovascular risk factors established in various clinical studies. We also review detailed experimental studies about the effect of bilirubin on atherosclerotic processes. In vitro, animal and human studies have proved that bilirubin inhibits oxidation of cholesterol which is an important step of atherosclerosis. Bilirubin attenuates chemotactic activity of monocytes and strongly inhibits adhesion of leukocytes to venule and production of pro-inflammatory cytokines. Bilirubin has inhibited serum-driven smooth muscle cell cycle progression at the G1 phase. Lastly, we will discuss briefly the influence of bilirubin on lipoprotein composition and endothelial dysfunction.
Collapse
Affiliation(s)
- Seung Joo Kang
- Department of Internal Medicine, Healthcare Research Institute, Seoul National University, Hospital Healthcare System Gangnam Center , Seoul , Korea
| | | | | |
Collapse
|
37
|
Langhans C, Weber-Carstens S, Schmidt F, Hamati J, Kny M, Zhu X, Wollersheim T, Koch S, Krebs M, Schulz H, Lodka D, Saar K, Labeit S, Spies C, Hubner N, Spranger J, Spuler S, Boschmann M, Dittmar G, Butler-Browne G, Mouly V, Fielitz J. Inflammation-induced acute phase response in skeletal muscle and critical illness myopathy. PLoS One 2014; 9:e92048. [PMID: 24651840 PMCID: PMC3961297 DOI: 10.1371/journal.pone.0092048] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 02/17/2014] [Indexed: 12/29/2022] Open
Abstract
Objectives Systemic inflammation is a major risk factor for critical-illness myopathy (CIM) but its pathogenic role in muscle is uncertain. We observed that interleukin 6 (IL-6) and serum amyloid A1 (SAA1) expression was upregulated in muscle of critically ill patients. To test the relevance of these responses we assessed inflammation and acute-phase response at early and late time points in muscle of patients at risk for CIM. Design Prospective observational clinical study and prospective animal trial. Setting Two intensive care units (ICU) and research laboratory. Patients/Subjects 33 patients with Sequential Organ Failure Assessment scores ≥8 on 3 consecutive days within 5 days in ICU were investigated. A subgroup analysis of 12 patients with, and 18 patients without CIM (non-CIM) was performed. Two consecutive biopsies from vastus lateralis were obtained at median days 5 and 15, early and late time points. Controls were 5 healthy subjects undergoing elective orthopedic surgery. A septic mouse model and cultured myoblasts were used for mechanistic analyses. Measurements and Main Results Early SAA1 expression was significantly higher in skeletal muscle of CIM compared to non-CIM patients. Immunohistochemistry showed SAA1 accumulations in muscle of CIM patients at the early time point, which resolved later. SAA1 expression was induced by IL-6 and tumor necrosis factor-alpha in human and mouse myocytes in vitro. Inflammation-induced muscular SAA1 accumulation was reproduced in a sepsis mouse model. Conclusions Skeletal muscle contributes to general inflammation and acute-phase response in CIM patients. Muscular SAA1 could be important for CIM pathogenesis. Trial Registration ISRCTN77569430.
Collapse
Affiliation(s)
- Claudia Langhans
- Experimental and Clinical Research Center (ECRC), a Cooperation between Max Delbrück Center and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Steffen Weber-Carstens
- Charité Universitätsmedizin Berlin, Campus Virchow and Campus Mitte, Anesthesiology and Operative Intensive Care Medicine, Berlin, Germany
| | - Franziska Schmidt
- Experimental and Clinical Research Center (ECRC), a Cooperation between Max Delbrück Center and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Jida Hamati
- Experimental and Clinical Research Center (ECRC), a Cooperation between Max Delbrück Center and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Melanie Kny
- Experimental and Clinical Research Center (ECRC), a Cooperation between Max Delbrück Center and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Xiaoxi Zhu
- Experimental and Clinical Research Center (ECRC), a Cooperation between Max Delbrück Center and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Tobias Wollersheim
- Charité Universitätsmedizin Berlin, Campus Virchow and Campus Mitte, Anesthesiology and Operative Intensive Care Medicine, Berlin, Germany
| | - Susanne Koch
- Charité Universitätsmedizin Berlin, Campus Virchow and Campus Mitte, Anesthesiology and Operative Intensive Care Medicine, Berlin, Germany
| | - Martin Krebs
- Charité Universitätsmedizin Berlin, Campus Virchow and Campus Mitte, Anesthesiology and Operative Intensive Care Medicine, Berlin, Germany
| | - Herbert Schulz
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Doerte Lodka
- Experimental and Clinical Research Center (ECRC), a Cooperation between Max Delbrück Center and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Kathrin Saar
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | | - Claudia Spies
- Charité Universitätsmedizin Berlin, Campus Virchow and Campus Mitte, Anesthesiology and Operative Intensive Care Medicine, Berlin, Germany
| | - Norbert Hubner
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Joachim Spranger
- Experimental and Clinical Research Center (ECRC), a Cooperation between Max Delbrück Center and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
- Charité Universitätsmedizin Berlin, NeuroCure Clinical Research Center, Berlin, Germany
- Charité Universitätsmedizin Berlin, Department of Endocrinology, Diabetes and Nutrition, Berlin, Germany
| | - Simone Spuler
- Experimental and Clinical Research Center (ECRC), a Cooperation between Max Delbrück Center and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Michael Boschmann
- Experimental and Clinical Research Center (ECRC), a Cooperation between Max Delbrück Center and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | - Gunnar Dittmar
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Gillian Butler-Browne
- Institut de Myologie, Institut national de la santé et de la recherche médicale, and L’Université Pierre et Marie Curie Paris, Paris, France
| | - Vincent Mouly
- Institut de Myologie, Institut national de la santé et de la recherche médicale, and L’Université Pierre et Marie Curie Paris, Paris, France
| | - Jens Fielitz
- Experimental and Clinical Research Center (ECRC), a Cooperation between Max Delbrück Center and Charité Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
- Charité Universitätsmedizin Berlin, Campus Virchow, Cardiology, Berlin, Germany
- * E-mail:
| |
Collapse
|
38
|
Yan S, Fu Q, Zhou Y, Zhang N, Zhou Q, Wang X, Yuan Z, Wang X, Du J, Zhang J, Zhan L. Establishment of stable reporter expression for in vivo imaging of nuclear factor-κB activation in mouse liver. Am J Cancer Res 2013; 3:841-50. [PMID: 24312154 PMCID: PMC3841335 DOI: 10.7150/thno.6997] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/16/2013] [Indexed: 12/18/2022] Open
Abstract
The nuclear factor-κB (NF-κB) signaling pathway plays a critical role in a multitude of cellular processes. Activation of the NF-κB transcription factor family is essential for the initiation of inflammation, immunity, cell proliferation and apoptosis through a list of responsive genes. In hepatic tissue, activation of the NF-κB pathway has been implicated in a number of pathological conditions. Here we described a mouse model for noninvasive quantification of NF-κB activation in the hepatic tissues. Mice were subjected to hydrodynamic delivery with a mixture of pattB-NF-κB-Fluc reporter and φC31o integrase vector. Hepatic expression of φC31o integrase mediated chromosomal integration of the pattB-NF-κB-Fluc reporter, resulting in stable luciferase expression at 300 days post transfection. We applied noninvasive imaging and were able to detect NF-κB activation under acute liver injury and hepatitis conditions. During hepatectomy-induced liver regeneration, NF-κB activation was detected locally in the tissues at the surgery site. Treatment with Sorafenib suppressed NF-κB activation, accompanied with perturbation of liver regeneration. In conclusion, we established a method for stable transfection of the hepatic tissues and applied the transfected mice to longitudinal monitoring of NF-κB activity under pathological conditions. Further exploration of this methodology for establishment of other disease models and for evaluation of novel pharmaceuticals is likely to be fruitful.
Collapse
|
39
|
Wright KT, Vella AT. RKIP contributes to IFN-γ synthesis by CD8+ T cells after serial TCR triggering in systemic inflammatory response syndrome. THE JOURNAL OF IMMUNOLOGY 2013; 191:708-16. [PMID: 23761631 DOI: 10.4049/jimmunol.1203486] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Systemic inflammatory response syndrome (SIRS) is associated with the development of severe medical complications, including progression to multiple organ dysfunction syndrome and even death. To date, only marginal improvements in terms of therapeutic options have been established for patients affected by SIRS. Raf kinase inhibitor protein (RKIP) is a regulator of MAPK and NF-κB signaling cascades, which are both critical for production of the proinflammatory cytokines responsible for SIRS initiation. By testing a T cell-dependent mouse model of SIRS that utilizes staphylococcal enterotoxin A specific for Vβ3(+) T cells, we show that RKIP is necessary for the exaggerated production of IFN-γ from SIRS splenocytes. This effect was not due to differences in T cell expansion, IL-10 production, or APC priming, but rather a cell-intrinsic defect lying downstream of the TCR in staphylococcal enterotoxin A-specific CD8(+) T cells. Importantly, mice lacking RKIP were still able to proliferate, survive, and contribute to cytokine production in response to pathogen associated molecular pattern-TLR-mediated stimuli, despite the TCR-dependent defects seen in our SIRS model. Finally, by blocking RKIP in wild-type SIRS splenocytes, the IFN-γ response by CD8(+) Vβ3(+) T cells was significantly diminished. These data suggest that RKIP may be a potential therapeutic target in SIRS by curbing effector cytokine production from CD8(+) T cells during serial TCR triggering.
Collapse
Affiliation(s)
- Kyle T Wright
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | |
Collapse
|
40
|
Rune I, Hansen CHF, Ellekilde M, Nielsen DS, Skovgaard K, Rolin BC, Lykkesfeldt J, Josefsen K, Tranberg B, Kihl P, Hansen AK. Ampicillin-improved glucose tolerance in diet-induced obese C57BL/6NTac mice is age dependent. J Diabetes Res 2013; 2013:319321. [PMID: 24369539 PMCID: PMC3863492 DOI: 10.1155/2013/319321] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 09/12/2013] [Accepted: 10/21/2013] [Indexed: 02/02/2023] Open
Abstract
Ampicillin has been shown to improve glucose tolerance in mice. We hypothesized that this effect is present only if treatment is initiated prior to weaning and that it disappears when treatment is terminated. High-fat fed C57BL/6NTac mice were divided into groups that received Ampicillin at different ages or not at all. We found that both diet and Ampicillin significantly changed the gut microbiota composition in the animals. Furthermore, there was a significant improvement in glucose tolerance in Ampicillin-treated, five-week-old mice compared to nontreated mice in the control group. At study termination, expressions of mRNA coding for tumor necrosis factor, serum amyloid A, and lactase were upregulated, while the expression of tumor necrosis factor (ligand) superfamily member 15 was downregulated in the ileum of Ampicillin-treated mice. Higher dendritic cell percentages were found systemically in high-fat diet mice, and a lower tolerogenic dendritic cell percentage was found both in relation to high-fat diet and late Ampicillin treatment. The results support our hypothesis that a "window" exists early in life in which an alteration of the gut microbiota affects glucose tolerance as well as development of gut immunity and that this window may disappear after weaning.
Collapse
Affiliation(s)
- I. Rune
- Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1870 Frederiksberg, Denmark
- *I. Rune:
| | - C. H. F. Hansen
- Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1870 Frederiksberg, Denmark
| | - M. Ellekilde
- Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1870 Frederiksberg, Denmark
| | - D. S. Nielsen
- Department of Food Science, Faculty of Science, University of Copenhagen, 1958 Frederiksberg, Denmark
| | - K. Skovgaard
- Innate Immunology Group, National Veterinary Institute, Technical University of Denmark, Bülowsvej 27, 1870 Frederiksberg, Denmark
| | - B. C. Rolin
- Translational Pharmacology, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - J. Lykkesfeldt
- Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1870 Frederiksberg, Denmark
| | - K. Josefsen
- The Bartholin Institute, Rigshospitalet Department 3733, Copenhagen Biocenter, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
| | - B. Tranberg
- Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1870 Frederiksberg, Denmark
| | - P. Kihl
- Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1870 Frederiksberg, Denmark
| | - A. K. Hansen
- Section of Experimental Animal Models, Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Thorvaldsensvej 57, 1870 Frederiksberg, Denmark
| |
Collapse
|
41
|
Karakas S, Mortada R, Fellow C. In search of the “LINK”: Acute Phase Serum Amyloid A. Atherosclerosis 2011; 216:266-8. [DOI: 10.1016/j.atherosclerosis.2011.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 01/25/2011] [Indexed: 11/15/2022]
|
42
|
Lopez de Heredia L, Gengatharan A, Foster J, Mather S, Magoulas C. Bioluminescence imaging of the brain response to acute inflammation in living C/EBP reporter mice. Neurosci Lett 2011; 497:134-8. [PMID: 21545825 DOI: 10.1016/j.neulet.2011.04.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 04/04/2011] [Accepted: 04/19/2011] [Indexed: 12/12/2022]
Abstract
The transcription factor CCAAT enhancer binding protein (C/EBP) is a key regulator of inflammation and immune responses, and recent studies suggest it is involved in inflammatory processes in the nervous system. We generated a transgenic reporter mouse model, carrying the luciferase (luc) gene under the transcriptional control of C/EBP, for visualising C/EBP activity in vivo. Real-time bioluminescence imaging reflecting C/EBP activity was performed in an acute inflammation model, after systemic administration of lipopolysaccharides (LPS), in C/EBP-luc mice. A striking activity of C/EBP was imaged predominantly in the brain of living C/EBP-luc mice in response to LPS, showing for the first time in vivo that C/EBP mediates the brain response to inflammation. Furthermore, dexamethasone, a potent anti-inflammatory agent, diminished the LPS-induced C/EBP activity demonstrating the physiological regulation of bioluminescence intensity in the brain of C/EBP-luc mice. Our results implicate that C/EBP reporter mice have the potential to be a valuable tool for studies on the mechanisms of brain inflammation in vivo and for the noninvasive preclinical evaluation of therapeutic agents targeting neuroinflammatory diseases.
Collapse
Affiliation(s)
- Luis Lopez de Heredia
- Centre for Neuroscience and Trauma, Blizard Institute of Cell and Molecular Science, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, 4 Newark Street, Whitechapel, London E12AT, UK
| | | | | | | | | |
Collapse
|
43
|
Sejima T, Kajiwara D, Kikuchi H, Imayoshi S, Yamauchi T, Ichimura K. Experimentally induced eosinophilic polyps in rabbit sinuses. Am J Rhinol Allergy 2011; 24:341-7. [PMID: 21244733 DOI: 10.2500/ajra.2010.24.3519] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Nasal polyps are one of the most common findings of physical examination in the otolaryngology area and the experimental model of nasal polyps in the rabbit maxillary sinus is helpful for clarifying the mechanism of polyp formation. Several protocols have been reported for this model, but most of them involved infectious polyps without eosinophil infiltration. We have attempted to establish a novel rabbit model of polyps associated with eosinophil infiltration. METHODS Rabbits were either untreated (group A) or sensitized with ovalbumin (OVA; groups B-D). After repeated exposure to OVA, some animals further received valine-glycine-serine-glutamine (group C) or poly-L-arginine (group D) in their maxillary sinuses for 4 weeks. Subsequently, sinus tissues were dissected and subjected to histological analysis. The changes in mRNA expression were analyzed by DNA microarray. RESULTS Remarkable histological changes were observed in groups C and D but not in group B in eosinophil number in the maxillary sinus mucosa, the width of the lamina propria, and polyp scoring. These changes in group D were greater than those in group C. DNA microarray analysis revealed that up-regulated genes in group D included those related to inflammation and extracellular matrix metabolism. On the other hand, down-regulated genes in group D involved those related to anti-inflammation. CONCLUSION Our results indicate that treatment with inflammatory agents, in combination with an antigen-dependent immune response, could induce nasal polyp formation associated with eosinophil infiltration and mucosal hypertrophy. The gene expression profile supported the clinical relevance of this model.
Collapse
Affiliation(s)
- Takayuki Sejima
- Department of Otorhinolaryngology–Head and Neck Surgery, Jichi Medical University School of Medicine, Tochigi, Japan.
| | | | | | | | | | | |
Collapse
|
44
|
Abstract
Conventional radiography, ultrasound, CT, MRI, and nuclear imaging are the current imaging modalities used for clinical evaluation of arthritis which is highly prevalent and a leading cause of disability. Some of these types of imaging are also used for monitoring disease progression and treatment response of arthritis. However, their disadvantages limit their utilities, such as ionizing radiation for radiography, CT, and nuclear imaging; suboptimal tissue contrast resolution for radiography, CT, ultrasound, and nuclear imaging; high cost for CT and MRI and nuclear imaging; and long data-acquisition time with ensuing patient discomfort for MRI. Recently, there have been considerable advances in nonionizing noninvasive optical imaging which has demonstrated promise for early diagnosis, monitoring therapeutic interventions and disease progression of arthritis. Optical based molecular imaging modalities such as fluorescence imaging have shown high sensitivity in detection of optical contrast agents and can aid early diagnosis and ongoing evaluation of chronic inflammatory arthritis. Optical transillumination imaging or diffuse optical tomography may differentiate normal joint clear synovial fluid from turbid and pink medium early in the inflammatory process. Fourier transform infrared spectroscopy has been used to evaluate fluid composition from joints affected by arthritis. Hemodynamic changes such as angiogenesis, hypervascularization, and hypoxia in arthritic articular tissue can potentially be observed by diffuse optical tomography and photoacoustic tomography. Optical measurements could also facilitate quantification of hemodynamic properties such as blood volume and oxygenation levels at early stages of inflammatory arthritis. Optical imaging provides methodologies which should contribute to detection of early changes and monitoring of progression in pathological characteristics of arthritis, with relatively simple instrumentation.
Collapse
Affiliation(s)
- David Chamberland
- Department of Radiology, University of Michigan School of Medicine, Ann Arbor, 48109, USA
| | | | | |
Collapse
|
45
|
Signore A, Mather SJ, Piaggio G, Malviya G, Dierckx RA. Molecular imaging of inflammation/infection: nuclear medicine and optical imaging agents and methods. Chem Rev 2010; 110:3112-45. [PMID: 20415479 DOI: 10.1021/cr900351r] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- A Signore
- Nuclear Medicine Unit, II Faculty of Medicine and Surgery, Sapienza University of Rome, Rome, Italy.
| | | | | | | | | |
Collapse
|
46
|
Kielland A, Carlsen H. Molecular imaging of transcriptional regulation during inflammation. JOURNAL OF INFLAMMATION-LONDON 2010; 7:20. [PMID: 20420665 PMCID: PMC2883981 DOI: 10.1186/1476-9255-7-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 04/26/2010] [Indexed: 01/21/2023]
Abstract
Molecular imaging enables non-invasive visualization of the dynamics of molecular processes within living organisms in vivo. Different imaging modalities as MRI, SPECT, PET and optic imaging are used together with molecular probes specific for the biological process of interest. Molecular imaging of transcription factor activity is done in animal models and mostly in transgenic reporter mice, where the transgene essentially consists of a promoter that regulates a reporter gene. During inflammation, the transcription factor NF-κB is widely involved in orchestration and regulation of the immune system and almost all imaging studies in this field has revolved around the role and regulation of NF-κB. We here present a brief introduction to experimental use and design of transgenic reporter mice and a more extensive review of the various studies where molecular imaging of transcriptional regulation has been applied during inflammation.
Collapse
Affiliation(s)
- Anders Kielland
- Dept, of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, PO Box 1046 Blindern, 0316 Oslo, Norway.
| | | |
Collapse
|
47
|
Tilahun AY, Theuer JE, Patel R, David CS, Rajagopalan G. Detrimental effect of the proteasome inhibitor, bortezomib in bacterial superantigen- and lipopolysaccharide-induced systemic inflammation. Mol Ther 2010; 18:1143-54. [PMID: 20372109 DOI: 10.1038/mt.2010.53] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Bacterial superantigen (BSAg)-induced toxic shock syndrome (TSS) and bacterial lipopolysaccharide (LPS)-induced shock are characterized by severe systemic inflammation. As nuclear factor kappaB (NF kappaB) plays an important role in inflammation and bortezomib, a proteasome inhibitor widely used in cancer chemotherapy, is a potent inhibitor of NF kappaB activation, we evaluated the therapeutic and prophylactic use of bortezomib in these conditions using murine models. Bortezomib prophylaxis significantly reduced serum levels of many cytokines and chemokines induced by BSAg. However, at 3 hours, serum level of TNF-a, an important cytokine implicated in TSS, was significantly reduced but not abolished. At 6 hours, there was no difference in the serum TNF-a levels between bortezomib treated and untreated mice challenged with staphylococcal enterotoxin B (SEB). Paradoxically, all mice treated with bortezomib either before or after BSAg challenge succumbed to TSS. Neither bortezomib nor BSAg was lethal if given alone. Serum biochemical parameters and histopathological findings suggested acute liver failure as the possible cause of mortality. Liver tissue from SEB-challenged mice treated with bortezomib showed a significant reduction in NF kappaB activation. Because NF kappaB-dependent antiapoptotic pathways protect hepatocytes from TNF-alpha-induced cell death, inhibition of NF kappaB brought forth by bortezomib in the face of elevated TNF-alpha levels caused by BSAg or LPS is detrimental.
Collapse
Affiliation(s)
- Ashenafi Y Tilahun
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
48
|
Transfusion of red blood cells after prolonged storage produces harmful effects that are mediated by iron and inflammation. Blood 2010; 115:4284-92. [PMID: 20299509 DOI: 10.1182/blood-2009-10-245001] [Citation(s) in RCA: 379] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although red blood cell (RBC) transfusions can be lifesaving, they are not without risk. In critically ill patients, RBC transfusions are associated with increased morbidity and mortality, which may increase with prolonged RBC storage before transfusion. The mechanisms responsible remain unknown. We hypothesized that acute clearance of a subset of damaged, stored RBCs delivers large amounts of iron to the monocyte/macrophage system, inducing inflammation. To test this in a well-controlled setting, we used a murine RBC storage and transfusion model to show that the transfusion of stored RBCs, or washed stored RBCs, increases plasma nontransferrin bound iron (NTBI), produces acute tissue iron deposition, and initiates inflammation. In contrast, the transfusion of fresh RBCs, or the infusion of stored RBC-derived supernatant, ghosts, or stroma-free lysate, does not produce these effects. Furthermore, the insult induced by transfusion of stored RBC synergizes with subclinical endotoxinemia producing clinically overt signs and symptoms. The increased plasma NTBI also enhances bacterial growth in vitro. Taken together, these results suggest that, in a mouse model, the cellular component of leukoreduced, stored RBC units contributes to the harmful effects of RBC transfusion that occur after prolonged storage. Nonetheless, these findings must be confirmed by prospective human studies.
Collapse
|
49
|
Liu J, Tang H, Niu L, Xu Y. Upregulation of Tanis mRNA expression in the liver is associated with insulin resistance in rats. TOHOKU J EXP MED 2010; 219:307-10. [PMID: 19966530 DOI: 10.1620/tjem.219.307] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Tanis, a newly discovered membrane protein, has been suggested to be involved in the development of insulin resistance (IR). In addition, Tanis was postulated as a hepatic receptor for an inflammatory marker, serum amyloid A-1 (SAA1), which represents a risk factor for cardiovascular disease. The aim of this study was to investigate the potential role of Tanis and SAA1 in the development of IR using a rat model. Male Sprague-Dawley (6 weeks old) rats were divided into three groups (n = 10 for each group): control, IR, and rosiglitazone-treated IR. Rosiglitazone is an insulin sensitizer that is used for treatment of type 2 diabetes. Rats were fed a high-fat diet for 35 days to induce IR, as judged by increased plasma glucose and insulin concentrations, following an oral glucose tolerance test. Rats of three groups were sacrificed, and the livers were dissected for RNA preparation. Real-time PCR analysis revealed that Tanis and SAA1 mRNA levels were significantly higher in the livers of IR rats than those of control rats fed a standard diet and those of IR rats treated for 35 days with rosiglitazone. Importantly, there was no significant difference in Tanis and SAA1 mRNA levels between control and rosiglitazone-treated IR rats. Furthermore, the expression levels of Tanis mRNA are positively correlated with those of SAA1 mRNA in the livers of control, IR and rosiglitazone-treated IR rats. In conclusion, high expression of Tanis mRNA in the liver is associated with the development of IR probably via the function of SAA1.
Collapse
Affiliation(s)
- Jie Liu
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | | | | | | |
Collapse
|
50
|
Reumann MK, Weiser MC, Mayer-Kuckuk P. Musculoskeletal molecular imaging: a comprehensive overview. Trends Biotechnol 2010; 28:93-101. [PMID: 20045210 DOI: 10.1016/j.tibtech.2009.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 10/26/2009] [Accepted: 11/12/2009] [Indexed: 01/01/2023]
Abstract
Molecular imaging permits non-invasive visualization and measurement of molecular and cell biology in living subjects, thereby complementing conventional anatomical imaging. Herein, we review the emerging application of molecular imaging for the study of musculoskeletal biology. Utilizing mainly bioluminescence and fluorescence techniques, molecular imaging has enabled in-vivo studies of (i) the activity of osteoblasts, osteoclasts, and hormones, (ii) the mechanisms of pathological cartilage and bone destruction, (iii) skeletal gene and cell therapy with and without biomaterial support, and (iv) the cellular processes in osteolysis and osteomyelitis. In these applications, musculoskeletal molecular imaging demonstrated feasibility for research in a myriad of musculoskeletal conditions ranging from bone fracture and arthritis to skeletal cancer. Importantly, these advances herald great potential for innovative clinical imaging in orthopedics, rheumatology, and oncology.
Collapse
Affiliation(s)
- Marie K Reumann
- Bone Cell Biology and Imaging Laboratory, Caspary Research Building, Rm. 623, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
| | | | | |
Collapse
|