51
|
Grądzki Z, Jarosz Ł, Stępień-Pyśniak D, Marek A. The effect of feed supplementation with Transcarpathian zeolite (clinoptilolite) on the concentrations of acute phase proteins and cytokines in the serum and hepatic tissue of chickens. Poult Sci 2020; 99:2424-2437. [PMID: 32359577 PMCID: PMC7597453 DOI: 10.1016/j.psj.2020.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/20/2019] [Accepted: 01/08/2020] [Indexed: 11/05/2022] Open
Abstract
The aim of the this study was to determine the effect of different levels of Transcarpathian zeolite (clinoptilolite) on selected indicators of the immune response in chickens by assessing the concentrations of the acute phase proteins haptoglobin, C-reactive protein (CRP), serum amyloid A, transferrin, and alpha-1-acid glycoprotein and the cytokines tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), IL-2, and IL-10 in the serum and hepatic tissues of chickens. The study was conducted on 450 one-day-old male Ross 308 chickens. The total rearing period was 42 D. The samples of blood and liver were collected before the start of the study (day 0) and on day 42, after rearing was completed. ELISA kits specific for chicken CRP, haptoglobin, alpha-1-acid glycoprotein, serum amyloid A, transferrin, TNF-α, IFN-γ, IL-2, and IL-10 were used to determine the levels of acute phase proteins and cytokines in the serum and liver homogenates. The results of immunological tests suggest that for long-term maintenance of homeostasis in chickens, the addition of 2% zeolite as a feed additive is most beneficial. The results indicate that 3% clinoptilolite induce production of Th1 pro-inflammatory cytokines, increasing the synthesis of IL-2, IFN-γ, and TNF-α. The high concentration of IL-10 after the use of zeolite in conjunction with the high concentration of IL-2, TNF-α, and IFN-γ indicates a reduction in the intensity of inflammatory processes, the enhancement of the humoral immune response, and the simultaneous inhibition of the production of Th1-type cytokines. The increase of CRP concentration in conjunction with high concentrations of pro- and anti-inflammatory cytokines in the birds from the group receiving 3% clinoptilolite demonstrates indicates that it can influence the development of local inflammatory processes and enhance immune regulation in birds. Our research has shown that clinoptilolite influences on an increase in birds' resistance to infection, as confirmed by clinical observations and anatomopathological examination and by the increase in the synthesis of acute phase proteins with immunoregulatory properties.
Collapse
Affiliation(s)
- Zbigniew Grądzki
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin 20-612, Poland
| | - Łukasz Jarosz
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin 20-612, Poland.
| | - Dagmara Stępień-Pyśniak
- Department of Veterinary Prevention and Avian Diseases, Faculty of Veterinary Medicine, Institute of Biological Bases of Animal Diseases, University of Life Sciences in Lublin, Lublin 20-612, Poland
| | - Agnieszka Marek
- Department of Veterinary Prevention and Avian Diseases, Faculty of Veterinary Medicine, Institute of Biological Bases of Animal Diseases, University of Life Sciences in Lublin, Lublin 20-612, Poland
| |
Collapse
|
52
|
Woodman C, Vundu G, George A, Wilson CM. Applications and strategies in nanodiagnosis and nanotherapy in lung cancer. Semin Cancer Biol 2020; 69:349-364. [PMID: 32088362 DOI: 10.1016/j.semcancer.2020.02.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 01/24/2020] [Accepted: 02/11/2020] [Indexed: 12/24/2022]
Abstract
Lung cancer is the second most common cancer and the leading cause of death in both men and women in the world. Lung cancer is heterogeneous in nature and diagnosis is often at an advanced stage as it develops silently in the lung and is frequently associated with high mortality rates. Despite the advances made in understanding the biology of lung cancer, progress in early diagnosis, cancer therapy modalities and considering the mechanisms of drug resistance, the prognosis and outcome still remains low for many patients. Nanotechnology is one of the fastest growing areas of research that can solve many biological problems such as cancer. A growing number of therapies based on using nanoparticles (NPs) have successfully entered the clinic to treat pain, cancer, and infectious diseases. Recent progress in nanotechnology has been encouraging and directed to developing novel nanoparticles that can be one step ahead of the cancer reducing the possibility of multi-drug resistance. Nanomedicine using NPs is continuingly impacting cancer diagnosis and treatment. Chemotherapy is often associated with limited targeting to the tumor, side effects and low solubility that leads to insufficient drug reaching the tumor. Overcoming these drawbacks of chemotherapy by equipping NPs with theranostic capability which is leading to the development of novel strategies. This review provides a synopsis of current progress in theranostic applications for lung cancer diagnosis and therapy using NPs including liposome, polymeric NPs, quantum dots, gold NPs, dendrimers, carbon nanotubes and magnetic NPs.
Collapse
Affiliation(s)
- Christopher Woodman
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, United Kingdom
| | - Gugulethu Vundu
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, United Kingdom
| | - Alex George
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, United Kingdom; Jubilee Centre for Medical Research, Jubilee Mission Medical College & Research Institute, Thrissur, Kerala, India
| | - Cornelia M Wilson
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, United Kingdom; University of Liverpool, Institute of Translation Medicine, Dept of Molecular & Clinical Cancer Medicine, United Kingdom; Novel Global Community Educational Foundation, Australia.
| |
Collapse
|
53
|
Wilfred BS, Madathil SK, Cardiff K, Urankar S, Yang X, Hwang HM, Gilsdorf JS, Shear DA, Leung LY. Alterations in Peripheral Organs following Combined Hypoxemia and Hemorrhagic Shock in a Rat Model of Penetrating Ballistic-Like Brain Injury. J Neurotrauma 2020; 37:656-664. [PMID: 31595817 PMCID: PMC7045350 DOI: 10.1089/neu.2019.6570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Polytrauma, with combined traumatic brain injury (TBI) and systemic damage are common among military and civilians. However, the pathophysiology of peripheral organs following polytrauma is poorly understood. Using a rat model of TBI combined with hypoxemia and hemorrhagic shock, we studied the status of peripheral redox systems, liver glycogen content, creatinine clearance, and systemic inflammation. Male Sprague-Dawley rats were subjected to hypoxemia and hemorrhagic shock insults (HH), penetrating ballistic-like brain injury (PBBI) alone, or PBBI followed by hypoxemia and hemorrhagic shock (PHH). Sham rats received craniotomy only. Biofluids and liver, kidney, and heart tissues were collected at 1 day, 2 days, 7 days, 14 days, and 28 days post-injury (DPI). Creatinine levels were measured in both serum and urine. Glutathione levels, glycogen content, and superoxide dismutase (SOD) and cytochrome C oxidase enzyme activities were quantified in the peripheral organs. Acute inflammation marker serum amyloid A-1 (SAA-1) level was quantified using western blot analysis. Urine to serum creatinine ratio in PHH group was significantly elevated on 7-28 DPI. Polytrauma induced a delayed disruption of the hepatic GSH/GSSG ratio, which resolved within 2 weeks post-injury. A modest decrease in kidney SOD activity was observed at 2 weeks after polytrauma. However, neither PBBI alone nor polytrauma changed the mitochondrial cytochrome C oxidase activity. Hepatic glycogen levels were reduced acutely following polytrauma. Acute inflammation marker SAA-1 showed a significant increase at early time-points following both systemic and brain injury. Overall, our findings demonstrate temporal cytological/tissue level damage to the peripheral organs due to combined PBBI and systemic injury.
Collapse
Affiliation(s)
- Bernard S Wilfred
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland
| | - Sindhu K Madathil
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland
| | - Katherine Cardiff
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland
| | - Sarah Urankar
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland
| | - Xiaofang Yang
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland
| | - Hye Mee Hwang
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland
| | - Janice S Gilsdorf
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland
| | - Deborah A Shear
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland
| | - Lai Yee Leung
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland.,Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
54
|
Skibiszewska S, Żaczek S, Dybala-Defratyka A, Jędrzejewska K, Jankowska E. Influence of short peptides with aromatic amino acid residues on aggregation properties of serum amyloid A and its fragments. Arch Biochem Biophys 2020; 681:108264. [PMID: 31945312 DOI: 10.1016/j.abb.2020.108264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/18/2019] [Accepted: 01/10/2020] [Indexed: 12/20/2022]
Abstract
Serum amyloid A variant 1.1 (SAA1.1) is an acute phase protein. In response to injury, inflammation or infection its production increases highly, which may lead to aggregation of the protein and accumulation of its deposits in various organs. Due to the cellular toxicity of the aggregates, as well as the fact that accumulated deposits are a burden that obstructs proper functioning of the affected tissues, it is vital to find a way to suppress the process of pathological aggregates formation. To make this possible, it is necessary to investigate thoroughly the oligomerization process and recognize factors that may influence its course. Some previous studies showed that aromatic interactions are important to the potential of an inhibitor to suppress the aggregation process. In our research we had proved that a five-residue peptide RSFFS (saa1-5) is an efficient inhibitor of aggregation of the most amyloidogenic fragment of SAA1.1, SAA1-12. In the present work the oligomerization and aggregation propensity of SAA1-12 was compared to that of SAA1-27, in order to determine the contribution of the sequence which extends beyond the most amyloidogenic region but encompasses residues reportedly involved in the stabilization of the SAA native conformation. Thioflavin T fluorescence assay, quantitative chromatographic analysis of the insoluble fraction and transmission electron microscopy allowed for a deeper insight into the SAA aggregation process and the morphology of aggregates. Substitutions of Phe3 and/or Phe4 residues in saa1-5 sequence with tryptophan, tyrosine, homophenylalanine, naphthylalanine and β,β-diphenylalanine allowed to study the influence of different aromatic systems on the aggregation of SAA1-12 and SAA1-27, and evaluate these results in relation to hSAA1.1 protein. Our results indicate that compounds with aromatic moieties can affect the course of the aggregation process and change the ratio between the soluble and insoluble aggregates.
Collapse
Affiliation(s)
- Sandra Skibiszewska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Szymon Żaczek
- Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924, Łódź, Poland
| | - Agnieszka Dybala-Defratyka
- Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924, Łódź, Poland
| | - Katarzyna Jędrzejewska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Elżbieta Jankowska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308, Gdańsk, Poland.
| |
Collapse
|
55
|
Hoyer FF, Naxerova K, Schloss MJ, Hulsmans M, Nair AV, Dutta P, Calcagno DM, Herisson F, Anzai A, Sun Y, Wojtkiewicz G, Rohde D, Frodermann V, Vandoorne K, Courties G, Iwamoto Y, Garris CS, Williams DL, Breton S, Brown D, Whalen M, Libby P, Pittet MJ, King KR, Weissleder R, Swirski FK, Nahrendorf M. Tissue-Specific Macrophage Responses to Remote Injury Impact the Outcome of Subsequent Local Immune Challenge. Immunity 2019; 51:899-914.e7. [PMID: 31732166 DOI: 10.1016/j.immuni.2019.10.010] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 09/06/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022]
Abstract
Myocardial infarction, stroke, and sepsis trigger systemic inflammation and organism-wide complications that are difficult to manage. Here, we examined the contribution of macrophages residing in vital organs to the systemic response after these injuries. We generated a comprehensive catalog of changes in macrophage number, origin, and gene expression in the heart, brain, liver, kidney, and lung of mice with myocardial infarction, stroke, or sepsis. Predominantly fueled by heightened local proliferation, tissue macrophage numbers increased systemically. Macrophages in the same organ responded similarly to different injuries by altering expression of tissue-specific gene sets. Preceding myocardial infarction improved survival of subsequent pneumonia due to enhanced bacterial clearance, which was caused by IFNɣ priming of alveolar macrophages. Conversely, EGF receptor signaling in macrophages exacerbated inflammatory lung injury. Our data suggest that local injury activates macrophages in remote organs and that targeting macrophages could improve resilience against systemic complications following myocardial infarction, stroke, and sepsis.
Collapse
Affiliation(s)
- Friedrich Felix Hoyer
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Kamila Naxerova
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Maximilian J Schloss
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Maarten Hulsmans
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Anil V Nair
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA; Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, BST 1720.1, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - David M Calcagno
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fanny Herisson
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Atsushi Anzai
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Yuan Sun
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Gregory Wojtkiewicz
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - David Rohde
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Vanessa Frodermann
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Katrien Vandoorne
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Gabriel Courties
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Christopher S Garris
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - David L Williams
- Department of Surgery and Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, 178 Maple Avenue, Johnson City, TN 37614, USA
| | - Sylvie Breton
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA; Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Dennis Brown
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA; Program in Membrane Biology and Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Michael Whalen
- Neuroscience Center and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, 55 Fruit Street, MA 02114, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Mikael J Pittet
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Kevin R King
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Division of Cardiology, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Ralph Weissleder
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | - Filip K Swirski
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA; Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA; Department of Internal Medicine I, University Hospital Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
56
|
Vietri L, Fui A, Bergantini L, d'Alessandro M, Cameli P, Sestini P, Rottoli P, Bargagli E. Serum amyloid A: A potential biomarker of lung disorders. Respir Investig 2019; 58:21-27. [PMID: 31708467 DOI: 10.1016/j.resinv.2019.09.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/29/2019] [Accepted: 09/20/2019] [Indexed: 12/27/2022]
Abstract
Serum amyloid A is an acute-phase protein with multiple immunological functions. Serum amyloid A is involved in lipid metabolism, inflammatory reactions, granuloma formation, and cancerogenesis. Additionally, serum amyloid A is involved in the pathogenesis of different autoimmune lung diseases. The levels of serum amyloid A has been evaluated in biological fluids of patients with different lung diseases, including autoimmune disorders, chronic obstructive pulmonary diseases, obstructive sleep apnea syndrome, sarcoidosis, asthma, lung cancer, and other lung disorders, such as idiopathic pulmonary fibrosis, tuberculosis, radiation pneumonitis, and cystic fibrosis. This review focuses on the cellular and molecular interactions of serum amyloid A in different lung diseases and suggests this acute-phase protein as a prognostic marker.
Collapse
Affiliation(s)
- Lucia Vietri
- Department of Medical and Surgical Sciences and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, Siena, Italy.
| | - Annalisa Fui
- Department of Medical and Surgical Sciences and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, Siena, Italy.
| | - Laura Bergantini
- Department of Medical and Surgical Sciences and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, Siena, Italy.
| | - Miriana d'Alessandro
- Department of Medical and Surgical Sciences and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, Siena, Italy.
| | - Paolo Cameli
- Department of Medical and Surgical Sciences and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, Siena, Italy.
| | - Piersante Sestini
- Department of Medical and Surgical Sciences and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, Siena, Italy.
| | - Paola Rottoli
- Department of Medical and Surgical Sciences and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, Siena, Italy.
| | - Elena Bargagli
- Department of Medical and Surgical Sciences and Neurosciences, Respiratory Disease and Lung Transplant Unit, Siena University, Siena, Italy.
| |
Collapse
|
57
|
Inflammation and TGF-β Signaling Differ between Abdominal Aneurysms and Occlusive Disease. J Cardiovasc Dev Dis 2019; 6:jcdd6040038. [PMID: 31683995 PMCID: PMC6955744 DOI: 10.3390/jcdd6040038] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/17/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023] Open
Abstract
Abdominal aortic aneurysms (AAA), are usually asymptomatic until rupture causes fatal bleeding, posing a major vascular health problem. AAAs are associated with advanced age, male gender, and cardiovascular risk factors (e.g. hypertension and smoking). Strikingly, AAA and AOD (arterial occlusive disease) patients have a similar atherosclerotic burden, yet develop either arterial dilatation or occlusion, respectively. The molecular mechanisms underlying this diversion are yet unknown. As this knowledge could improve AAA treatment strategies, we aimed to identify genes and signaling pathways involved. We compared RNA expression profiles of abdominal aortic AAA and AOD patient samples. Based on differential gene expression profiles, we selected a gene set that could serve as blood biomarker or as pharmacological intervention target for AAA. In this AAA gene list we identified previously AAA-associated genes COL11A1, ADIPOQ, and LPL, thus validating our approach as well as novel genes; CXCL13, SLC7A5, FDC-SP not previously linked to aneurysmal disease. Pathway analysis revealed overrepresentation of significantly altered immune-related pathways between AAA and AOD. Additionally, we found bone morphogenetic protein (BMP) signaling inhibition simultaneous with activation of transforming growth factor β (TGF-β) signaling associated with AAA. Concluding our gene expression profiling approach identifies novel genes and an interplay between BMP and TGF-β signaling regulation specifically for AAA.
Collapse
|
58
|
Al Saleh AS, Sidiqi MH, Gertz MA. Improvement in Gastrointestinal Symptoms From Light Chain Amyloidosis After Adalimumab Therapy. Mayo Clin Proc 2019; 94:1380-1381. [PMID: 31272581 DOI: 10.1016/j.mayocp.2019.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/09/2019] [Accepted: 04/25/2019] [Indexed: 10/26/2022]
Affiliation(s)
- Abdullah S Al Saleh
- Mayo Clinic, Rochester, MN; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | | | | |
Collapse
|
59
|
Evidence for the important role of inflammation in xenotransplantation. JOURNAL OF INFLAMMATION-LONDON 2019; 16:10. [PMID: 31148951 PMCID: PMC6537172 DOI: 10.1186/s12950-019-0213-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
There is increasing evidence of a sustained state of systemic inflammation after pig-to-nonhuman primate (NHP) xenotransplantation (that has been termed systemic inflammation in xenograft recipients [SIXR]). Increases in inflammatory markers, e.g., C-reactive protein, histones, serum amyloid A, D-dimer, cytokines, chemokines, and a decrease in free triiodothyronine, have been demonstrated in the recipient NHPs. The complex interactions between inflammation, coagulation, and the immune response are well-recognized, but the role of inflammation in xenograft recipients is not fully understood. The evidence suggests that inflammation can promote the activation of coagulation and the adaptive immune response, but the exact mechanisms remain uncertain. If prolonged xenograft survival is to be achieved, anti-inflammatory strategies (e.g., the administration of anti-inflammatory agents, and/or the generation of genetically-engineered organ-source pigs that are protected from the effect of inflammation) may be necessary to prevent, control, or negate the effect of the systemic inflammation that develops in xenograft recipients. This may allow for a reduction in the intensity of exogenous immunosuppressive therapy. If immunological tolerance to a xenograft is to be obtained, then control of inflammation may be essential.
Collapse
|
60
|
Wang Y, Zhang Y, Chi X, Ma X, Xu W, Shi F, Hu S. Anti-inflammatory mechanism of ginsenoside Rg1: Proteomic analysis of milk from goats with mastitis induced with lipopolysaccharide. Int Immunopharmacol 2019; 71:382-391. [PMID: 30952102 DOI: 10.1016/j.intimp.2019.03.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/23/2019] [Accepted: 03/25/2019] [Indexed: 12/20/2022]
Abstract
Previous investigation showed that intravenous injection of ginsenoside Rg1 had a therapeutic effect on Escherichia coli lipopolysaccharide-induced mastitis in lactating goats and it protected animals from lipopolysaccharide challenge via toll-like receptor 4 signaling pathway. The present study was to use proteomic approach to explore the anti-inflammatory mechanisms of Rg1. Nine dairy goats were randomly divided into three groups with 3 animals in each: groups 1 and 2 received intra-mammary infusion of lipopolysaccharide and then intravenously injected with saline or Rg1 solution; animals in group 3 were first intramammarily and then intravenously administered saline solution, and served as a control group. Milk whey at 6 h post lipopolysaccharide challenge was prepared for tandem mass tags based quantitative proteomic analysis. The results showed that 791 proteins were totally identified from the whey. Of them, 98 proteins between groups 1 (lipopolysaccharide + Saline) and 3 (Saline + Saline), and 34 proteins between groups 2 (lipopolysaccharide + Rg1) and 1 were significantly different. Group 1 than group 3 had significantly more inflammatory factors such as interleukin 6, acute phase proteins, blood coagulation factors, complement proteins, and oxidative stress markers while these factors were reduced in group 2 treated with Rg1. In addition, proteins in group 2 associated with peroxisome-proliferator-activated receptor γ activation and recovery of milk fat and production were upregulated compared to group 1. Therefore, Rg1 may exert its anti-inflammatory effect on lipopolysaccharide-induced mastitis in goats via modulating expression of proteins relating to peroxisome-proliferator-activated receptor γ and toll-like receptor 4 signaling pathway.
Collapse
Affiliation(s)
- Yuemin Wang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hang Zhou 310058, PR China.
| | - Yong Zhang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hang Zhou 310058, PR China
| | - Xiaoqing Chi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hang Zhou 310058, PR China
| | - Xiaodan Ma
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hang Zhou 310058, PR China
| | - Wei Xu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hang Zhou 310058, PR China
| | - Fushan Shi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hang Zhou 310058, PR China
| | - Songhua Hu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hang Zhou 310058, PR China.
| |
Collapse
|
61
|
Griffiths K, Maxwell AP, McCarter RV, Nicol P, Hogg RE, Harbinson M, McKay GJ. Serum amyloid A levels are associated with polymorphic variants in the serum amyloid A 1 and 2 genes. Ir J Med Sci 2019; 188:1175-1183. [PMID: 30852808 DOI: 10.1007/s11845-019-01996-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/22/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Serum amyloid A (SAA) is secreted by liver hepatocytes in response to increased inflammation whereupon it associates with high-density lipoprotein (HDL) and alters the protein and lipid composition of HDL negating some of its anti-atherogenic properties. AIMS To identify variants within the SAA gene that may be associated with SAA levels and/or cardiovascular disease (CVD). METHODS We identified exonic variants within the SAA genes by deoxyribonucleic acid (DNA) Sanger sequencing. We tested the association between SAA variants and serum SAA levels in 246 individuals with and without CVD. RESULTS Increased SAA was associated with rs2468844 (beta [β] = 1.73; confidence intervals [CI], 1.14-1.75; p = 0.01), rs1136747 (β = 1.53 (CI, 1.11-1.73); p = 0.01) and rs149926073 (β = 3.37 (CI, 1.70-4.00); p = 0.02), while rs1136745 was significantly associated with decreased SAA levels (β = 0.70 (CI, 0.53-0.94); p = 0.02). Homozygous individuals with the SAA1.3 haplotype had significantly lower levels of SAA compared with those with SAA1.1 or SAA1.5 (β = 0.43 (CI, 0.22-0.85); p = 0.02) while SAA1.3/1.5 heterozygotes had significantly higher SAA levels compared with those homozygous for SAA1.1 (β = 2.58 (CI, 1.19-5.57); p = 0.02). CONCLUSIONS We have identified novel genetic variants in the SAA genes associated with SAA levels, a biomarker of inflammation and chronic disease. The utility of SAA as a biomarker for inflammation and chronic disease may be influenced by underlying genetic variation in baseline levels.
Collapse
Affiliation(s)
- Kayleigh Griffiths
- Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland
| | - Alexander P Maxwell
- Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland
| | - Rachel V McCarter
- Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland
| | - Patrick Nicol
- Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland
| | - Ruth E Hogg
- Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland
| | - Mark Harbinson
- Centre for Medical Education, Queen's University Belfast, Belfast, Northern Ireland
- Department of Cardiology, Belfast Health and Social Care Trust, Royal Hospital, Belfast, Northern Ireland
| | - Gareth J McKay
- Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland.
- Centre for Public Health, Institute of Clinical Sciences, Block B, Royal Victoria Hospital, Queen's University Belfast, Belfast, BT12 6BA, Ireland.
| |
Collapse
|
62
|
Chami B, Hossain F, Hambly TW, Cai X, Aran R, Fong G, Vellajo A, Martin NJJ, Wang X, Dennis JM, Sharma A, Shihata WA, Chin-Dusting JPF, de Haan JB, Sharland A, Geczy CL, Freedman B, Witting PK. Serum Amyloid A Stimulates Vascular and Renal Dysfunction in Apolipoprotein E-Deficient Mice Fed a Normal Chow Diet. Front Immunol 2019; 10:380. [PMID: 30899260 PMCID: PMC6416175 DOI: 10.3389/fimmu.2019.00380] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/14/2019] [Indexed: 12/19/2022] Open
Abstract
Elevated serum amyloid A (SAA) levels may promote endothelial dysfunction, which is linked to cardiovascular and renal pathologies. We investigated the effect of SAA on vascular and renal function in apolipoprotein E-deficient (ApoE−/−) mice. Male ApoE−/− mice received vehicle (control), low-level lipopolysaccharide (LPS), or recombinant human SAA by i.p. injection every third day for 2 weeks. Heart, aorta and kidney were harvested between 3 days and 18 weeks after treatment. SAA administration increased vascular cell adhesion molecule (VCAM)-1 expression and circulating monocyte chemotactic protein (MCP)-1 and decreased aortic cyclic guanosine monophosphate (cGMP), consistent with SAA inhibiting nitric oxide bioactivity. In addition, binding of labeled leukocytes to excised aorta increased as monitored using an ex vivo leukocyte adhesion assay. Renal injury was evident 4 weeks after commencement of SAA treatment, manifesting as increased plasma urea, urinary protein, oxidized lipids, urinary kidney injury molecule (KIM)-1 and multiple cytokines and chemokines in kidney tissue, relative to controls. Phosphorylation of nuclear-factor-kappa-beta (NFκB-p-P65), tissue factor (TF), and macrophage recruitment increased in kidneys from ApoE−/− mice 4 weeks after SAA treatment, confirming that SAA elicited a pro-inflammatory and pro-thrombotic phenotype. These data indicate that SAA impairs endothelial and renal function in ApoE−/− mice in the absence of a high-fat diet.
Collapse
Affiliation(s)
- Belal Chami
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Farjaneh Hossain
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Thomas W Hambly
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Xiaoping Cai
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Roshanak Aran
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Genevieve Fong
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Abigail Vellajo
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Nathan J J Martin
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - XiaoSuo Wang
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Joanne M Dennis
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Arpeeta Sharma
- Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Waled A Shihata
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jaye P F Chin-Dusting
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.,Department of Pharmacology, Monash University, Melbourne, VIC, Australia.,Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Judy B de Haan
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Alexandra Sharland
- Transplantation Immunobiology Group, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Carolyn L Geczy
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Ben Freedman
- ANZAC Research and Heart Research Institutes, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Paul K Witting
- Discipline of Pathology, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
63
|
Murdoch CC, Espenschied ST, Matty MA, Mueller O, Tobin DM, Rawls JF. Intestinal Serum amyloid A suppresses systemic neutrophil activation and bactericidal activity in response to microbiota colonization. PLoS Pathog 2019; 15:e1007381. [PMID: 30845179 PMCID: PMC6405052 DOI: 10.1371/journal.ppat.1007381] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 01/09/2019] [Indexed: 12/25/2022] Open
Abstract
The intestinal microbiota influences the development and function of myeloid lineages such as neutrophils, but the underlying molecular mechanisms are unresolved. Using gnotobiotic zebrafish, we identified the immune effector Serum amyloid A (Saa) as one of the most highly induced transcripts in digestive tissues following microbiota colonization. Saa is a conserved secreted protein produced in the intestine and liver with described effects on neutrophils in vitro, however its in vivo functions remain poorly defined. We engineered saa mutant zebrafish to test requirements for Saa on innate immunity in vivo. Zebrafish mutant for saa displayed impaired neutrophil responses to wounding but augmented clearance of pathogenic bacteria. At baseline, saa mutants exhibited moderate neutrophilia and altered neutrophil tissue distribution. Molecular and functional analyses of isolated neutrophils revealed that Saa suppresses expression of pro-inflammatory markers and bactericidal activity. Saa's effects on neutrophils depended on microbiota colonization, suggesting this protein mediates the microbiota's effects on host innate immunity. To test tissue-specific roles of Saa on neutrophil function, we over-expressed saa in the intestine or liver and found that sufficient to partially complement neutrophil phenotypes observed in saa mutants. These results indicate Saa produced by the intestine in response to microbiota serves as a systemic signal to neutrophils to restrict aberrant activation, decreasing inflammatory tone and bacterial killing potential while simultaneously enhancing their ability to migrate to wounds.
Collapse
Affiliation(s)
- Caitlin C. Murdoch
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Scott T. Espenschied
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Molly A. Matty
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Olaf Mueller
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - David M. Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| |
Collapse
|
64
|
Lee JW, Stone ML, Porrett PM, Thomas SK, Komar CA, Li JH, Delman D, Graham K, Gladney WL, Hua X, Black TA, Chien AL, Majmundar KS, Thompson JC, Yee SS, O'Hara MH, Aggarwal C, Xin D, Shaked A, Gao M, Liu D, Borad MJ, Ramanathan RK, Carpenter EL, Ji A, de Beer MC, de Beer FC, Webb NR, Beatty GL. Hepatocytes direct the formation of a pro-metastatic niche in the liver. Nature 2019; 567:249-252. [PMID: 30842658 PMCID: PMC6430113 DOI: 10.1038/s41586-019-1004-y] [Citation(s) in RCA: 257] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/12/2019] [Indexed: 12/25/2022]
Abstract
The liver is the most common site of metastatic disease1. Although this metastatic tropism may reflect the mechanical trapping of circulating tumour cells, liver metastasis is also dependent, at least in part, on the formation of a 'pro-metastatic' niche that supports the spread of tumour cells to the liver2,3. The mechanisms that direct the formation of this niche are poorly understood. Here we show that hepatocytes coordinate myeloid cell accumulation and fibrosis within the liver and, in doing so, increase the susceptibility of the liver to metastatic seeding and outgrowth. During early pancreatic tumorigenesis in mice, hepatocytes show activation of signal transducer and activator of transcription 3 (STAT3) signalling and increased production of serum amyloid A1 and A2 (referred to collectively as SAA). Overexpression of SAA by hepatocytes also occurs in patients with pancreatic and colorectal cancers that have metastasized to the liver, and many patients with locally advanced and metastatic disease show increases in circulating SAA. Activation of STAT3 in hepatocytes and the subsequent production of SAA depend on the release of interleukin 6 (IL-6) into the circulation by non-malignant cells. Genetic ablation or blockade of components of IL-6-STAT3-SAA signalling prevents the establishment of a pro-metastatic niche and inhibits liver metastasis. Our data identify an intercellular network underpinned by hepatocytes that forms the basis of a pro-metastatic niche in the liver, and identify new therapeutic targets.
Collapse
Affiliation(s)
- Jae W Lee
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Meredith L Stone
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paige M Porrett
- Division of Transplant Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stacy K Thomas
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chad A Komar
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joey H Li
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Devora Delman
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kathleen Graham
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Whitney L Gladney
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xia Hua
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Taylor A Black
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Austin L Chien
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Krishna S Majmundar
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey C Thompson
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephanie S Yee
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark H O'Hara
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charu Aggarwal
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dong Xin
- Division of Transplant Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Abraham Shaked
- Division of Transplant Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mingming Gao
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Dexi Liu
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, USA
| | - Mitesh J Borad
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ, USA
| | - Ramesh K Ramanathan
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ, USA
- Merck Research Labs, Rahway, NJ, USA
| | - Erica L Carpenter
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ailing Ji
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Maria C de Beer
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Frederick C de Beer
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
| | - Nancy R Webb
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, USA
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Gregory L Beatty
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
65
|
Involvement of serum amyloid A1 in the rupture of fetal membranes through induction of collagen I degradation. Clin Sci (Lond) 2019; 133:515-530. [PMID: 30683734 DOI: 10.1042/cs20180950] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/03/2019] [Accepted: 01/25/2019] [Indexed: 12/22/2022]
Abstract
The de novo synthesis of serum amyloid A1 (SAA1) is augmented in human fetal membranes at parturition. However, its role in parturition remains largely unknown. Here, we investigated whether SAA1 was involved in the rupture of fetal membranes, a crucial event in parturition accompanied with extensive degradation of collagens. Results showed that SAA1 decreased both intracellular and extracellular COL1A1 and COL1A2 abundance, the two subunits of collagen I, without affecting their mRNA levels in human amnion fibroblasts. These reductions were completely blocked only with inhibition of both matrix metalloproteases (MMPs) and autophagy. Consistently, SAA1 increased MMP-2/9 abundance and the markers for autophagic activation including autophagy related (ATG) 7 (ATG7) and the microtubule-associated protein light chain 3 β (LC3B) II/I ratio with the formation of LC3 punctas and autophagic vacuoles in the fibroblasts. Moreover, the autophagic degradation of COL1A1/COL1A2 and activation of MMP-2/9 by SAA1 were blocked by inhibitors for the toll-like receptors 2/4 (TLR2/4) or NF-κB. Finally, reciprocal corresponding changes of SAA1 and collagen I were observed in the amnion following spontaneous rupture of membranes (ROM) at parturition. Conclusively, SAA1 may participate in membrane rupture at parturition by degradating collagen I via both autophagic and MMP pathways. These effects of SAA1 appear to be mediated by the TLR2/4 receptors and the NF-κB pathway.
Collapse
|
66
|
Kurvits L, Reimann E, Kadastik-Eerme L, Truu L, Kingo K, Erm T, Kõks S, Taba P, Planken A. Serum Amyloid Alpha Is Downregulated in Peripheral Tissues of Parkinson's Disease Patients. Front Neurosci 2019; 13:13. [PMID: 30760975 PMCID: PMC6361740 DOI: 10.3389/fnins.2019.00013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 01/08/2019] [Indexed: 11/13/2022] Open
Abstract
We report the changed levels of serum amyloid alpha, an immunologically active protein, in Parkinson’s disease (PD) patients’ peripheral tissues. We have previously shown that Saa-1 and -2 (serum amyloid alpha-1,-2, genes) were among the top downregulated genes in PD patients’ skin, using whole-genome RNA sequencing. In the current study, we characterized the gene and protein expression profiles of skin and blood samples from patients with confirmed PD diagnosis and age/sex matched controls. qRT-PCR analysis of PD skin demonstrated downregulation of Saa-1 and -2 genes in PD patients. However, the lowered amount of protein could not be visualized using immunohistochemistry, due to low quantity of SAA (Serum Amyloid Alpha, protein) in skin. Saa-1 and -2 expression levels in whole blood were below detection threshold based on RNA sequencing, however significantly lowered protein levels of SAA1/2 in PD patients’ serum were shown with ELISA, implying that SAA is secreted into the blood. These results show that SAA is differentially expressed in the peripheral tissues of PD patients.
Collapse
Affiliation(s)
- Lille Kurvits
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia.,Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ene Reimann
- Institute of Pathophysiology, University of Tartu, Tartu, Estonia
| | - Liis Kadastik-Eerme
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia
| | - Laura Truu
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Külli Kingo
- Department of Dermatology, University of Tartu, Tartu, Estonia.,Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Triin Erm
- Department of Pathology, Tartu University Hospital, Tartu, Estonia
| | - Sulev Kõks
- Centre for Comparative Genomics, Murdoch University, Perth, WA, Australia.,Perron Institute for Neurological and Translational Science, University of Western Australia, Perth, WA, Australia
| | - Pille Taba
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia
| | - Anu Planken
- Oncology and Haematology Clinic, North-Estonian Medical Centre, Tallinn, Estonia
| |
Collapse
|
67
|
Ignacio RMC, Gibbs CR, Kim S, Lee ES, Adunyah SE, Son DS. Serum amyloid A predisposes inflammatory tumor microenvironment in triple negative breast cancer. Oncotarget 2019; 10:511-526. [PMID: 30728901 PMCID: PMC6355188 DOI: 10.18632/oncotarget.26566] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/29/2018] [Indexed: 12/20/2022] Open
Abstract
Acute-phase proteins (APPs) are associated with a variety of disorders such as infection, inflammatory diseases, and cancers. The signature profile of APPs in breast cancer (BC) is poorly understood. Here, we identified serum amyloid A (SAA) for proinflammatory predisposition in BC through the signature profiles of APPs, interleukin (IL) and tumor necrosis factor (TNF) superfamily using publicly available datasets of tumor samples and cell lines. Triple-negative breast cancer (TNBC) subtype highly expressed SAA1/2 compared to HER2, luminal A (LA) and luminal B (LB) subtypes. IL1A, IL1B, IL8/CXCL8, IL32 and IL27RA in IL superfamily and CD70, TNFSF9 and TNFRSF21 in TNF superfamily were highly expressed in TNBC compared to other subtypes. SAA is restrictedly regulated by nuclear factor (NF)-κB and IL-1β, an NF-κB activator highly expressed in TNBC, increased the promoter activity of SAA1 in human TNBC MDA-MB231 cells. Interestingly, two κB-sites contained in SAA1 promoter were involved, and the proximal region (-96/-87) was more critical than the distal site (-288/-279) in regulating IL-1β-induced SAA1. Among the SAA receptors, TLR1 and TLR2 were highly expressed in TNBC. Cu-CPT22, TLR1/2 antagonist, abrogated IL-1β-induced SAA1 promoter activity. In addition, SAA1 induced IL8/CXCL8 promoter activity, which was partially reduced by Cu-CPT22. Notably, SAA1/2, TLR2 and IL8/CXCL8 were associated with a poor overall survival in mesenchymal-like TNBC. Taken together, IL-1-induced SAA via NF-κB-mediated signaling could potentiate an inflammatory burden, leading to cancer progression and high mortality in TNBC patients.
Collapse
Affiliation(s)
- Rosa Mistica C Ignacio
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| | - Carla R Gibbs
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| | - Soohyun Kim
- Department of Veterinary Sciences, College of Veterinary Medicine, Kon-Kuk University, Seoul, Republic of Korea
| | - Eun-Sook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, USA
| | - Samuel E Adunyah
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| | - Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN, USA
| |
Collapse
|
68
|
Jarosz Ł, Marek A, Grądzki Z, Laskowska E, Kwiecień M. Effect of Zinc Sulfate and Zinc Glycine Chelate on Concentrations of Acute Phase Proteins in Chicken Serum and Liver Tissue. Biol Trace Elem Res 2019; 187:258-272. [PMID: 29675569 PMCID: PMC6314988 DOI: 10.1007/s12011-018-1346-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/10/2018] [Indexed: 01/13/2023]
Abstract
The aim of the study was to determine how inorganic and organic forms of zinc affect the concentrations of C-reactive protein (CRP), serum amyloid A (SAA), alpha-1-acid glycoprotein (α-1-AGP), haptoglobin (Hp), and transferrin (TRF) in the blood and liver tissue of 450 1-day-old Ross 308 chicken. Four experimental groups received one the following: inorganic zinc (ZnSO4), a zinc phytase enzyme supplement (ZnSO4-F), organic zinc in combination with glycine (Zn-Gly), or organic zinc supplemented with phytase (Zn-Gly-F). The chicken serum and liver homogenates were assayed using an ELISA kit. The results of the study showed statistically significantly higher serum and liver concentration of SAA in the group of birds that received zinc sulfate in comparison to the group of birds receiving zinc in organic form. A statistically significantly higher serum concentration of CRP and α-1-AGP was also noted in the group receiving zinc sulfate as compared to the Zn-Gly group. Comparison of the serum concentration of TRF between the supplemented groups showed a statistically significant increase in this parameter in the Zn-Gly-F group as compared to the ZSO4-F group. The increase in the serum concentration of Hp in all groups in comparison to the control may indicate stimulation of local immune mechanisms. The results of this study showed an increase in the concentrations of APPs such as AGP and TRF following the administration of zinc glycine chelates, which may demonstrate their effect on metabolic processes in the liver and on immunocompetent cells that regulate the intensity of the immune response.
Collapse
Affiliation(s)
- Łukasz Jarosz
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka 30, 20-612 Lublin, Poland
| | - Agnieszka Marek
- Sub-Department of Preventive Veterinary and Avian Diseases, Institute of Biological Bases of Animal Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka 30, 20-612 Lublin, Poland
| | - Zbigniew Grądzki
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka 30, 20-612 Lublin, Poland
| | - Ewa Laskowska
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Głęboka 30, 20-612 Lublin, Poland
| | - Małgorzata Kwiecień
- Faculty of Biology and Animal Breeding, Institute of Animal Nutrition and Bromatology, Department of Animal Nutrition, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland
| |
Collapse
|
69
|
Singh RD, Shandilya R, Bhargava A, Kumar R, Tiwari R, Chaudhury K, Srivastava RK, Goryacheva IY, Mishra PK. Quantum Dot Based Nano-Biosensors for Detection of Circulating Cell Free miRNAs in Lung Carcinogenesis: From Biology to Clinical Translation. Front Genet 2018; 9:616. [PMID: 30574163 PMCID: PMC6291444 DOI: 10.3389/fgene.2018.00616] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 11/23/2018] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is the most frequently occurring malignancy and the leading cause of cancer-related death for men in our country. The only recommended screening method is clinic based low-dose computed tomography (also called a low-dose CT scan, or LDCT). However, the effect of LDCT on overall mortality observed in lung cancer patients is not statistically significant. Over-diagnosis, excessive cost, risks associated with radiation exposure, false positive results and delay in the commencement of the treatment procedure questions the use of LDCT as a reliable technique for population-based screening. Therefore, identification of minimal-invasive biomarkers able to detect malignancies at an early stage might be useful to reduce the disease burden. Circulating nucleic acids are emerging as important source of information for several chronic pathologies including lung cancer. Of these, circulating cell free miRNAs are reported to be closely associated with the clinical outcome of lung cancer patients. Smaller size, sequence homology between species, low concentration and stability are some of the major challenges involved in characterization and specific detection of miRNAs. To circumvent these problems, synthesis of a quantum dot based nano-biosensor might assist in sensitive, specific and cost-effective detection of differentially regulated miRNAs. The wide excitation and narrow emission spectra of these nanoparticles result in excellent fluorescent quantum yields with a broader color spectrum which make them ideal bio-entities for fluorescence resonance energy transfer (FRET) based detection for sequential or simultaneous study of multiple targets. In addition, photo-resistance and higher stability of these nanoparticles allows extensive exposure and offer state-of-the art sensitivity for miRNA targeting. A major obstacle for integrating QDs into clinical application is the QD-associated toxicity. However, the use of non-toxic shells along with surface modification not only overcomes the toxicity issues, but also increases the ability of QDs to quickly detect circulating cell free miRNAs in a non-invasive mode. The present review illustrates the importance of circulating miRNAs in lung cancer diagnosis and highlights the translational prospects of developing QD-based nano-biosensor for rapid early disease detection.
Collapse
Affiliation(s)
- Radha D. Singh
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Ruchita Shandilya
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Arpit Bhargava
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Rajat Kumar
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Rajnarayan Tiwari
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, India
| | - Rupesh K. Srivastava
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Irina Y. Goryacheva
- Department of General and Inorganic Chemistry, Saratov State University, Saratov, Russia
| | - Pradyumna K. Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| |
Collapse
|
70
|
Kilic L, Erden A, Sener YZ, Armagan B, Sari A, Kalyoncu U, Karadag O, Akdogan A, Dogan I, Apras Bilgen S, Kiraz S, Ertenli I. Rituximab Therapy in Renal Amyloidosis Secondary to Rheumatoid Arthritis. Biomolecules 2018; 8:136. [PMID: 30400666 PMCID: PMC6316109 DOI: 10.3390/biom8040136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/17/2018] [Accepted: 10/24/2018] [Indexed: 12/03/2022] Open
Abstract
Secondary amyloid A (AA) amyloidosis is a late and serious complication of poorly controlled, chronic inflammatory diseases. Rheumatoid arthritis (RA) patients with poorly controlled, longstanding disease and those with extra-articular manifestations are under risk for the development of AA amyloidosis. Although new drugs have proven to be significantly effective in the treatment of secondary AA amyloidosis, no treatment modality has proven to be ideal. To date, only in small case series preliminary clinical improvement have been shown with rituximab therapy for AA amyloidosis secondary to RA that is refractory to TNF-α inhibitors (TNF-i) therapy. In these case series, we assessed the efficacy and safety of rituximab therapy for patients with RA and secondary amyloidosis. Hacettepe University Biologic Registry was developed at 2005. The data of the RA patients who were prescribed a biological drug were recorded regularly. Patients with biopsy proven AA amyloidosis patients were screened. Of 1022 RA patients under biologic therapy, 0.7% patients had clinically apparent histologically confirmed amyloidosis. Four of seven patients who were prescribed rituximab at least one infusion enrolled to those case series. Two of four patients showed significant clinical improvement and one of them also had decrease in proteinuria and the other one had stable renal function and proteinuria. The main goal for the treatment of AA amyloidosis is to control the activity of the underlying disorder. In this study, we showed that rituximab may be an effective treatment in RA patients with amyloidosis who were unresponsive to conventional disease modifying anti-rheumatic drugs (DMARDs) and/or TNFi.
Collapse
Affiliation(s)
- Levent Kilic
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| | - Abdulsamet Erden
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| | - Yusuf Ziya Sener
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| | - Berkan Armagan
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| | - Alper Sari
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| | - Umut Kalyoncu
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| | - Omer Karadag
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| | - Ali Akdogan
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| | - Ismail Dogan
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| | - Sule Apras Bilgen
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| | - Sedat Kiraz
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| | - Ihsan Ertenli
- Department of Internal Medicine, Division of Rheumatology, Hacettepe University Faculty of Medicine, 06100 Ankara, Turkey.
| |
Collapse
|
71
|
Lucherini OM, Lopalco G, Cantarini L, Emmi G, Lopalco A, Venerito V, Vitale A, Iannone F. Critical regulation of Th17 cell differentiation by serum amyloid-A signalling in Behcet's disease. Immunol Lett 2018; 201:38-44. [PMID: 30385329 DOI: 10.1016/j.imlet.2018.10.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/18/2018] [Accepted: 10/23/2018] [Indexed: 10/28/2022]
Abstract
The current understandings on cellular and molecular biology suggest that Th17 axis plays a pivotal role in Behcet's disease (BD) pathogenesis. Recently the role of serum amyloid-A (SAA) as a potential marker of disease activity in BD patients has been explored, and it has been reported that the occurrence of specific clinical features are significantly associated with high serum levels of this inflammatory mediator. The aim of this study was to investigate the cytokine-like activity of SAA in inducing Th17 differentiation from CD4 + T naive cells in BD. Purified peripheral monocytes from BD and healthy control (HC) were stimulated with SAA "in vitro", and secreted IL-8, TNF-α, IL-18, IFN-α, IFN-γ, IL-1β and IL-6 were measured using a Bio-Rad multiplex cytokine immunoassay. To assess Th17 differentiation, purified CD4 + T cells were challenged with anti-CD3/CD28 antibodies, while cultured with supernatant derived from SAA stimulated monocytes, and intracellular staining of IL-17A and IFN-γ was evaluated by flow-cytometry. Furthermore, peripheral blood mononuclear cells (PBMCs) were stimulated with SAA and transcript levels of RAR-related orphan nuclear receptor (ROR)-γt and IL-17A were assessed by Real-time PCR. Upon stimulation with SAA, monocytes obtained from both HC and BD groups released large amounts of IL-8, IL-6, TNF-α, IL-1β and IFN-α. Monocytes-derived supernatants from BD patients, but not HC, were capable of promoting Th17 but not Th1 differentiation from CD4 + T cells. However, SAA did not induce up-regulation of Th17 specific mRNA transcript such as IL-17A and (ROR)-γt in PBMCs from both HC and BD. In BD patients SAA induced Th17 polarization rather than Th1 differentiation from CD4 + T cells. These data suggest that a critical regulation of Th17 may be the functional link between acute SAA increase and the induction of Th17 mediated inflammatory response in BD.
Collapse
Affiliation(s)
- Orso Maria Lucherini
- Research Center of Systemic Autoinflammatory Diseases, Behçet's Disease Clinic and Rheumatology-Ophthalmology Collaborative Uveitis Center, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Giuseppe Lopalco
- Department of Emergency and Organ Transplantation, Rheumatology Unit, Bari, Italy.
| | - Luca Cantarini
- Research Center of Systemic Autoinflammatory Diseases, Behçet's Disease Clinic and Rheumatology-Ophthalmology Collaborative Uveitis Center, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, Italy
| | - Antonio Lopalco
- Department of Pharmacy-Drug Sciences, University of Bari, Bari, Italy
| | - Vincenzo Venerito
- Department of Emergency and Organ Transplantation, Rheumatology Unit, Bari, Italy
| | - Antonio Vitale
- Research Center of Systemic Autoinflammatory Diseases, Behçet's Disease Clinic and Rheumatology-Ophthalmology Collaborative Uveitis Center, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Florenzo Iannone
- Department of Emergency and Organ Transplantation, Rheumatology Unit, Bari, Italy
| |
Collapse
|
72
|
Sharma A, Govindan P, Toukatly M, Healy J, Henry C, Senter S, Najafian B, Kestenbaum B. Heroin Use Is Associated with AA-Type Kidney Amyloidosis in the Pacific Northwest. Clin J Am Soc Nephrol 2018; 13:1030-1036. [PMID: 29907621 PMCID: PMC6032593 DOI: 10.2215/cjn.13641217] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/17/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVES AA-type kidney amyloidosis is classically associated with chronic autoimmune or inflammatory disorders. However, some urban centers have reported a high prevalence of injection drug use among patients with kidney AA amyloidosis. Previous reports lack control groups to quantify associations and most predate the opioid epidemic in the United States. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We conducted a case-control study of 38 patients with biopsy-confirmed kidney AA amyloidosis and 72 matched control individuals without this condition from two large hospital systems in Seattle, Washington. We ascertained the pattern and duration of heroin use by medical chart review and determined associations using logistic regression. RESULTS Among case patients, 95% had a prior history of heroin use, 87% had skin abscesses, and 76% and 27% had evidence of muscling and skin popping, respectively. After adjustment for age, race, sex, site, and year of biopsy, any heroin use (past or current) was associated with an estimated 170-times higher risk of kidney AA amyloidosis compared with no heroin use (95% confidence interval, 28 to 1018 times higher; P<0.001). Chronic autoimmune disorders were uncommon among case patients in this study. The median time to ESKD among patients with AA amyloidosis was 2.4 years (interquartile range, 0.5-7.5 years). CONCLUSIONS Injection heroin use is strongly associated with kidney AA amyloidosis in the Pacific Northwest. Unique aspects of heroin use, in particular geographic regions or frequent associated soft-tissue infections, may be an important cause of this progressive kidney disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Steve Senter
- Institute for Translational Health Sciences, University of Washington, Seattle, Washington
| | | | - Bryan Kestenbaum
- Division of Nephrology, Department of Medicine
- Kidney Research Institute, and
| |
Collapse
|
73
|
Varan Ö, Kucuk H, Babaoglu H, Guven SC, Ozturk MA, Haznedaroglu S, Goker B, Tufan A. Efficacy and safety of interleukin-1 inhibitors in familial Mediterranean fever patients complicated with amyloidosis. Mod Rheumatol 2018; 29:363-366. [PMID: 29578360 DOI: 10.1080/14397595.2018.1457469] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Colchicine is the mainstay of the treatment of familial Mediterranean fever (FMF). However, 10% of FMF patients do not respond well to colchicine. Efficacy of interleukin (IL)-1 inhibitors in reducing attacks have been demonstrated in colchicine-resistant FMF (crFMF) patients recently. Colchicine is still the only approved drug for the prevention of amyloidosis in FMF and utility of IL-1 inhibitors in crFMF cases who already has amyloidosis remain to be elucidated. Herein, we evaluated efficacy and safety of IL-1 inhibitors in patients with crFMF-associated AA amyloidosis in a relatively large single center study. METHODS Medical records of FMF patients complicated with AA amyloidosis in our dedicated FMF center were retrospectively reviewed and those patients who ever treated with IL-1 inhibitors were enrolled into the study. Patient global, physician global assessments (on 0-10 cm visual analog scale), C-reactive protein (CRP), erythrocyte sedimentation rate (ESR), serum creatinine and 24-h urinary protein excretion values for each visit were recruited from computer-based hospital records. Treatment response of patients were assessed with clinical symptoms, serum albumin, CRP and ESR values. Renal outcome parameters were analyzed on those not receiving renal replacement therapy. RESULTS Seventeen patients were identified with crFMF-amyloidosis that ever treated with IL-1 inhibitors. Background colchicine therapy was continued in all patients in maximal-tolerated dose along with IL-1 inhibitors. All patients benefit from IL-1 antagonists assessed by patient and physician global assessments. Inflammatory markers, CRP and ESR, were significantly reduced in all and normalized in 12 out of 17 patients. More importantly, the amount of proteinuria was remarkably improved following IL-1 inhibitor therapy (1606 mg/day to 519 mg/day, p = .008). Both anakinra and canakinumab were well-tolerated without severe side effects. All patients were initially treated with anakinra but switched to canakinumab in seven patients (one leukopenia, four injection site reaction, two inefficacy). CONCLUSION We evaluated the clinical and laboratory responses to IL-1 inhibitors in crFMF-associated amyloidosis patients. We found significant decreases in CRP, ESR and proteinuria after IL-1 inhibitor therapy. This study confirmed that IL-1 inhibitors are effective for controlling attacks and inflammatory activity in FMF patients complicated with AA amyloidosis. Moreover, they reduce or stabilize amount of proteinuria and preserve renal function in short-term follow-up. Prolonged prospective clinical trials are warranted to assess their long-term efficacy in this particular patient group.
Collapse
Affiliation(s)
- Özkan Varan
- a Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology , Gazi University , Ankara , Turkey
| | - Hamit Kucuk
- a Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology , Gazi University , Ankara , Turkey
| | - Hakan Babaoglu
- a Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology , Gazi University , Ankara , Turkey
| | - Serdar Can Guven
- b Faculty of Medicine, Department of Physical Therapy and Rehabilitation, Division of Rheumatology , Gazi University , Ankara , Turkey
| | - Mehmet Akif Ozturk
- a Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology , Gazi University , Ankara , Turkey
| | - Seminur Haznedaroglu
- a Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology , Gazi University , Ankara , Turkey
| | - Berna Goker
- a Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology , Gazi University , Ankara , Turkey
| | - Abdurrahman Tufan
- a Faculty of Medicine, Department of Internal Medicine, Division of Rheumatology , Gazi University , Ankara , Turkey
| |
Collapse
|
74
|
Lane AP, Truong-Tran QA, Myers A, Bickel C, Schleimer RP. Serum Amyloid A, Properdin, Complement 3, and Toll-Like Receptors are Expressed Locally in Human Sinonasal Tissue. ACTA ACUST UNITED AC 2018. [DOI: 10.1177/194589240602000122] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background There is a growing appreciation of the role that nasal mucosa plays in innate immunity. In this study, the expression of pattern recognition receptors known as toll-like receptors (TLRs) and the effector molecules complement factor 3 (C3), properdin, and serum amyloid A (SAA) were examined in human sinonasal mucosa obtained from control subjects and patients with chronic rhinosinusitis (CRS). Methods Sinonasal mucosal specimens were obtained from 20 patients with CRS and 5 control subjects. Messenger RNA (mRNA) was isolated and tested using Taqman real-time polymerase chain reaction with primer and probe sets for C3, complement factor P, and SAA. Standard polymerase chain reaction was performed for the 10 known TLRs. Immunohistochemistry was performed on the microscopic sections using antibodies against C3 Results Analysis of the sinonasal sample mRNA revealed expression of all 10 TLRs in both CRS samples and in control specimens. Expression of the three effector proteins was detected also, with the levels of mRNA for C3 generally greater than SAA and properdin in CRS patients. No significant differences were found in TLR or innate immune protein expression in normal controls. Immunohistochemical analysis of sinonasal mucosal specimens established C3 staining ranging from 20 to 85% of the epithelium present. Conclusion These studies indicate that sinonasal mucosa expresses genes involved in innate immunity including the TLRs and proteins involved in complement activation. We hypothesize that local production of complement and acute phase proteins by airway epithelium on stimulation of innate immune receptors may play an important role in host defense in the airway and, potentially, in the pathogenesis of CRS.
Collapse
Affiliation(s)
- Andrew P. Lane
- Department of Otolaryngology–Head and Neck Surgery, Johns Hopkins, University School of Medicine, Baltimore, Maryland
| | - Quynh-Ai Truong-Tran
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Allan Myers
- Johns Hopkins, Bayview Asthma and Allergy Center, Baltimore, Maryland
| | - Carol Bickel
- Johns Hopkins, Bayview Asthma and Allergy Center, Baltimore, Maryland
| | - Robert P. Schleimer
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
75
|
Celiac Disease and Secondary Amyloidosis: A Possible Causal Association? ACG Case Rep J 2018; 5:e24. [PMID: 29619399 PMCID: PMC5876448 DOI: 10.14309/crj.2018.24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 02/02/2018] [Indexed: 02/04/2023] Open
Abstract
We report a rare case of secondary renal amyloidosis in a patient with probable celiac disease presenting as nephrotic syndrome. A 30-year-old man with chronic diarrhea presented with progressive anasarca for 2 years. On further evaluation, he had hypoalbuminemia, significant nephrotic-range proteinuria, and elevated levels of anti-tissue transglutaminase. Renal biopsy suggested deposition of amorphous Congo red–positive material in the glomerular mesangium, positive for amyloid A amyloidosis. Endoscopic duodenal biopsy suggested blunting of the villous architecture and chronic inflammation of the lamina propria. The patient subsequently developed massive pulmonary embolism and died due to refractory cardiogenic shock.
Collapse
|
76
|
Serum Amyloid A1 Is an Epithelial Prorestitutive Factor. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:937-949. [PMID: 29366677 DOI: 10.1016/j.ajpath.2017.12.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 11/10/2017] [Accepted: 12/05/2017] [Indexed: 12/12/2022]
Abstract
Several proteins endogenously produced during the process of intestinal wound healing have demonstrated prorestitutive properties. The presence of serum amyloid A1 (SAA1), an acute-phase reactant, within inflamed tissues, where it exerts chemotaxis of phagocytes, is well recognized; however, a putative role in intestinal wound repair has not been described. Herein, we show that SAA1 induces intestinal epithelial cell migration, spreading, and attachment through a formyl peptide receptor 2-dependent mechanism. Induction of the prorestitutive phenotype is concentration and time dependent and is associated with epithelial reactive oxygen species production and alterations in p130 Crk-associated substrate staining. In addition, using a murine model of wound recovery, we provide evidence that SAA1 is dynamically and temporally regulated, and that the elaboration of SAA1 within the wound microenvironment correlates with the influx of SAA1/CD11b coexpressing immune cells and increases in cytokines known to induce SAA expression. Overall, the present work demonstrates an important role for SAA in epithelial wound recovery and provides evidence for a physiological role in the wound environment.
Collapse
|
77
|
Shang J, Yamashita T, Fukui Y, Song D, Li X, Zhai Y, Nakano Y, Morihara R, Hishikawa N, Ohta Y, Abe K. Different Associations of Plasma Biomarkers in Alzheimer's Disease, Mild Cognitive Impairment, Vascular Dementia, and Ischemic Stroke. J Clin Neurol 2018; 14:29-34. [PMID: 29629537 PMCID: PMC5765253 DOI: 10.3988/jcn.2018.14.1.29] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 08/05/2017] [Accepted: 08/10/2017] [Indexed: 12/22/2022] Open
Abstract
Background and Purpose Cognitive and cerebrovascular diseases are common in the elderly, but differences in the plasma levels and associations of plasma biomarkers in these diseases remain elusive. Methods The present study investigated differences in plasma fatty acids [eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)], adiponectin, reptin, plasma markers of inflammation [high-sensitivity C-reactive protein (hsCRP) and serum amyloid A (serum AA)], and plasma lipids [high-density lipoprotein and low-density lipoprotein (LDL)] in patients with Alzheimer's disease (AD) (n=266), mild cognitive impairment (MCI) (n=44), vascular dementia (VaD) (n=33), and ischemic stroke (IS) (n=200) in comparison to normal controls (n=130). Results The serological data showed that lower EPA and DHA levels and higher reptin and LDL levels were associated with AD and IS, the reptin/adiponectin ratio was strongly associated with IS, the hsCRP level was more strongly associated with VaD and IS, and the serum AA level was associated with all three cognitive diseases and IS. Conclusions This is the first report of differences in the expression levels of plasma biomarkers and peripheral arterial tonometry among AD, MCI, VaD, and IS patients and normal controls. These different associations indicate that diverse pathological mechanisms underlie these diseases.
Collapse
Affiliation(s)
- Jingwei Shang
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Dongjing Song
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Xianghong Li
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yun Zhai
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yumiko Nakano
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Nozomi Hishikawa
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yasuyuki Ohta
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| |
Collapse
|
78
|
Yang M, Liu F, Higuchi K, Sawashita J, Fu X, Zhang L, Zhang L, Fu L, Tong Z, Higuchi K. Serum amyloid A expression in the breast cancer tissue is associated with poor prognosis. Oncotarget 2017; 7:35843-35852. [PMID: 27058895 PMCID: PMC5094967 DOI: 10.18632/oncotarget.8561] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 02/28/2016] [Indexed: 12/14/2022] Open
Abstract
Background Serum amyloid A (SAA), an acute-phase protein, is expressed primarily in the liver, and recently found also expressed in cancer tissues. However, its expression and prognostic value in breast cancer have not been described. Results SAA protein was found expressed in tumor cells in 44.2% cases and in TAM in 62.5% cases. FISH showed more frequent SAA mRNA expression in TAM than in tumor cells (76% versus 12%, p < 0.001), and a significant association between the frequencies of SAA mRNA expression in TAM and tumor cells (rs = 0.603, p < 0.001). The immunoreactivities of SAA protein in TAM and tumor cells were both associated with lymphovascular invasion and lymph node metastasis. Moreover, SAA-positivity in TAMs was associated with larger tumor-size, higher histological-grade, negative estrogen-receptor and progesterone-receptor statuses, and HER-2 overexpression. It was also linked to worse recurrence-free survival in a multivariable regression model. Methods Immunohistochemistry was applied on the tumor tissues from 208 breast cancer patients to evaluate the local SAA-protein expression with additional CD68 stain to identify the tumor-associated macrophage (TAM) on the serial tissue sections. Fluorescent in situ hybridization (FISH) was conducted on serial tissue sections from 25 of the 208 tumors to examine the expression and location of SAA mRNA. Conclusions Our results suggested that the TAMs may be a pivotal and main source of SAA production in tumor microenvironment of breast cancer. SAA immunoreactivity in TAM is associated with worse recurrence-free survival, and is therefore a biomarker candidate for postoperative surveillance and perhaps a therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Mu Yang
- Department of Aging Biology, Institute of Pathogenesis and Disease Prevention, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Fangfang Liu
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Kayoko Higuchi
- Department of Pathology, Aizawa Hospital, Matsumoto, Japan
| | - Jinko Sawashita
- Department of Aging Biology, Institute of Pathogenesis and Disease Prevention, Shinshu University Graduate School of Medicine, Matsumoto, Japan.,Department of Biological Sciences for Intractable Neurological Diseases, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Xiaoying Fu
- Department of Pathology, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Li Zhang
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lanjing Zhang
- Department of Pathology, University Medical Center of Princeton, Plainsboro, NJ, USA.,Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Department of Pathology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA.,Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Li Fu
- Department of Breast Pathology and Research Laboratory, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Zhongsheng Tong
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Keiichi Higuchi
- Department of Aging Biology, Institute of Pathogenesis and Disease Prevention, Shinshu University Graduate School of Medicine, Matsumoto, Japan.,Department of Biological Sciences for Intractable Neurological Diseases, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| |
Collapse
|
79
|
Van den Bossche L, Schoonenberg VAC, Burgener IA, Penning LC, Schrall IM, Kruitwagen HS, van Wolferen ME, Grinwis GCM, Kummeling A, Rothuizen J, van Velzen JF, Stathonikos N, Molenaar MR, Helms BJ, Brouwers JFHM, Spee B, van Steenbeek FG. Aberrant hepatic lipid storage and metabolism in canine portosystemic shunts. PLoS One 2017; 12:e0186491. [PMID: 29049355 PMCID: PMC5648188 DOI: 10.1371/journal.pone.0186491] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a poorly understood multifactorial pandemic disorder. One of the hallmarks of NAFLD, hepatic steatosis, is a common feature in canine congenital portosystemic shunts. The aim of this study was to gain detailed insight into the pathogenesis of steatosis in this large animal model. Hepatic lipid accumulation, gene-expression analysis and HPLC-MS of neutral lipids and phospholipids in extrahepatic (EHPSS) and intrahepatic portosystemic shunts (IHPSS) was compared to healthy control dogs. Liver organoids of diseased dogs and healthy control dogs were incubated with palmitic- and oleic-acid, and lipid accumulation was quantified using LD540. In histological slides of shunt livers, a 12-fold increase of lipid content was detected compared to the control dogs (EHPSS P<0.01; IHPSS P = 0.042). Involvement of lipid-related genes to steatosis in portosystemic shunting was corroborated using gene-expression profiling. Lipid analysis demonstrated different triglyceride composition and a shift towards short chain and omega-3 fatty acids in shunt versus healthy dogs, with no difference in lipid species composition between shunt types. All organoids showed a similar increase in triacylglycerols after free fatty acids enrichment. This study demonstrates that steatosis is probably secondary to canine portosystemic shunts. Unravelling the pathogenesis of this hepatic steatosis might contribute to a better understanding of steatosis in NAFLD.
Collapse
Affiliation(s)
- Lindsay Van den Bossche
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Vivien A. C. Schoonenberg
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Iwan A. Burgener
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Louis C. Penning
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Ingrid M. Schrall
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Hedwig S. Kruitwagen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Monique E. van Wolferen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Guy C. M. Grinwis
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Anne Kummeling
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jan Rothuizen
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jeroen F. van Velzen
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nikolas Stathonikos
- Department of Pathology, University Medical Center, Utrecht, The Netherlands
| | - Martijn R. Molenaar
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht, The Netherlands
| | - Bernd J. Helms
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht, The Netherlands
| | - Jos F. H. M. Brouwers
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine & Institute of Biomembranes, Utrecht, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Frank G. van Steenbeek
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
80
|
Mao JY, Sun JT, Yang K, Shen WF, Lu L, Zhang RY, Tong X, Liu Y. Serum amyloid A enrichment impairs the anti-inflammatory ability of HDL from diabetic nephropathy patients. J Diabetes Complications 2017; 31:1538-1543. [PMID: 28760652 DOI: 10.1016/j.jdiacomp.2017.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/06/2017] [Accepted: 07/07/2017] [Indexed: 01/13/2023]
Abstract
AIMS Impaired anti-inflammatory ability of high-density lipoprotein (HDL) has been demonstrated in patients with type-2 diabetes mellitus (T2DM). However, whether HDL from patients with diabetic nephropathy (DN) suffers additional damage remains unknown. This study compared the anti-inflammatory capacities of HDL from healthy controls, T2DM patients with normal renal function, and T2DM patients with DN. MATERIALS AND METHODS HDL was isolated from healthy controls (n=33) and T2DM patients with normal renal function (n=21), chronic kidney disease (CKD) (n=27), and end-stage renal disease (ESRD) (n=27). Human peripheral blood mononuclear cells (PBMCs) from healthy volunteers were pretreated with HDL (100μg/mL) for 1h, then incubated with lipopolysaccharide (LPS) (50ng/mL) for 24h. The anti-inflammatory ability of HDL was measured as the secretion of TNF-α in LPS-activated monocytes. RESULTS The anti-inflammatory ability of HDL was gradually impaired as kidney function declined. Serum amyloid A (SAA) concentration in HDLDN significantly increased and was positively correlated with the impaired anti-inflammatory ability of HDL (Pearson r=0.315, P=0.006). Furthermore, HDL supplemented with SAA significantly increased TNF-α release from PBMCs compared with that from control HDL. CONCLUSIONS These findings identified an impaired anti-inflammatory capacity of HDL from DN patients, which might be attributable to SAA enrichment.
Collapse
MESH Headings
- Adult
- Aged
- Cells, Cultured
- China/epidemiology
- Cross-Sectional Studies
- Diabetes Mellitus, Type 2/complications
- Diabetic Angiopathies/epidemiology
- Diabetic Angiopathies/immunology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Diabetic Nephropathies/immunology
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/pathology
- Diabetic Nephropathies/physiopathology
- Female
- Hospitals, University
- Humans
- Kidney/physiopathology
- Kidney Failure, Chronic/complications
- Kidney Failure, Chronic/immunology
- Kidney Failure, Chronic/metabolism
- Kidney Failure, Chronic/pathology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/pathology
- Lipopolysaccharides/toxicity
- Lipoproteins, HDL/blood
- Lipoproteins, HDL/isolation & purification
- Lipoproteins, HDL/metabolism
- Male
- Middle Aged
- Outpatient Clinics, Hospital
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/immunology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Risk Factors
- Serum Amyloid A Protein/analysis
- Serum Amyloid A Protein/metabolism
- Severity of Illness Index
- Vasculitis/complications
- Vasculitis/immunology
- Vasculitis/metabolism
- Vasculitis/pathology
Collapse
Affiliation(s)
- Jing Yan Mao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jia Teng Sun
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ke Yang
- Institute of Cardiovascular Disease, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wei Feng Shen
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Institute of Cardiovascular Disease, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lin Lu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rui Yan Zhang
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuemei Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yan Liu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
81
|
O'Reilly S. Toll Like Receptors in systemic sclerosis: An emerging target. Immunol Lett 2017; 195:2-8. [PMID: 28888416 DOI: 10.1016/j.imlet.2017.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/25/2017] [Accepted: 09/01/2017] [Indexed: 12/21/2022]
Abstract
Pattern Recognition Receptors are critical receptors that elicit an immune response upon their activation that culminates in activation of NF-KB and cytokine secretion. Key among these receptors are the Toll-Like Receptors (TLRs). These evolutionary conserved receptors form a key part in the defence against various pathogens and comprise a key part of the innate immune system. Systemic sclerosis is an autoimmune disease in which a breach of tolerance has occurred and leads to fulminant autoimmunity, dysregulated cytokines, pro-fibrotic mediators and activation of fibroblasts leading to fibrosis via collagen deposition. It has become apparent in recent years that the innate immune system and specifically TLRs are important in disease pathogenesis; responding to internal ligands to initiate an innate immune response ultimately leading to release of a variety of factors that initiate and perpetuate fibrosis. This review will examine the recent evidence of TLR signalling in systemic sclerosis and the internal danger associated molecules that may mediate the fibrotic cascade. Evaluation of their contribution to disease in systemic sclerosis and possible therapeutic targeting will be discussed.
Collapse
Affiliation(s)
- Steven O'Reilly
- Faculty of Health and Life Sciences, Northumbria University, Ellison Building, Newcastle Upon Tyne, United Kingdom.
| |
Collapse
|
82
|
Nair AM, Goel R, Hindhumati M, Jayakanthan K, Visalakshi J, Joseph G, Danda S, Danda D. Serum amyloid A as a marker of disease activity and treatment response in Takayasu arteritis. Rheumatol Int 2017; 37:1643-1649. [PMID: 28801814 DOI: 10.1007/s00296-017-3786-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/05/2017] [Indexed: 01/26/2023]
Abstract
Assessment of disease activity in Takayasu arteritis (TA) is challenging. We aimed to study utility of serum amyloid A (SAA) to assess disease activity and its association with SAA gene polymorphisms, if any, in our TA patients. Serum of 99 consecutive adult TA patients and 40 healthy controls were assayed for SAA. Depending on the ITAS2010 and ITAS-CRP score, patients were designated as having active disease if ITAS2010 ≥ 2 or ITAS-CRP ≥ 3 and stable disease if ITAS2010 = 0 or ITAS-CRP is ≤1. Clinical ITAS of 0 with raised inflammatory markers scoring a ITAS-CRP of 2 was considered as indeterminate for disease activity assessment. Repeat SAA levels for active group was measured after 6 months from baseline. SAA levels between active and stable disease as well as serial levels were compared. DNA of 40 patients and controls were genotyped for SAA polymorphisms (rs12218, rs2468844) and the allele frequencies were compared. At baseline, SAA levels were higher in patients as compared to controls (137.4 vs 100.8 ng/ml, p = 0.001) and higher in patients with active disease (166.4 ng/ml) than those with stable disease (98.2 ng/ml), p = 0.001. SAA decreased during follow-up in treatment responders (189.9 ng/ml at baseline vs 119.0 ng/ml at follow-up, p = 0.008); in contrast, there was no significant change among non-responders during follow-up. Allelic frequencies of SAA gene polymorphisms did not differ between cases and controls. SAA may be a reliable biomarker to assess disease activity and treatment response in TA.
Collapse
Affiliation(s)
- Aswin M Nair
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - Ruchika Goel
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - M Hindhumati
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - K Jayakanthan
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, 632004, India
| | - J Visalakshi
- Department of Biostatistics, Christian Medical College, Vellore, Tamil Nadu, India
| | - George Joseph
- Department of Cardiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Sumita Danda
- Department of Clinical Genetics, Christian Medical College, Vellore, Tamil Nadu, India
| | - Debashish Danda
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, 632004, India.
| |
Collapse
|
83
|
Frame NM, Jayaraman S, Gantz DL, Gursky O. Serum amyloid A self-assembles with phospholipids to form stable protein-rich nanoparticles with a distinct structure: A hypothetical function of SAA as a "molecular mop" in immune response. J Struct Biol 2017. [PMID: 28645735 DOI: 10.1016/j.jsb.2017.06.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Serum amyloid A (SAA) is an acute-phase protein whose action in innate immunity and lipid homeostasis is unclear. Most circulating SAA binds plasma high-density lipoproteins (HDL) and reroutes lipid transport. In vivo SAA binds existing lipoproteins or generates them de novo upon lipid uptake from cells. We explored the products of SAA-lipid interactions and lipoprotein remodeling in vitro. SAA complexes with palmitoyl-oleoyl phosphocholine (POPC) were analyzed for structure and stability using circular dichroism and fluorescence spectroscopy, electron microscopy, gel electrophoresis and gel filtration. The results revealed the formation of 8-11nm lipoproteins that were∼50% α-helical and stable at near-physiological conditions but were irreversibly remodeled at Tm∼52°C. Similar HDL-size nanoparticles formed spontaneously at ambient conditions or upon thermal remodeling of parent lipoproteins containing various amounts of proteins and lipids, including POPC and cholesterol. Therefore, such HDL-size particles formed stable kinetically accessible structures in a wide range of conditions. Based on their size and stoichiometry, each particle contained about 12 SAA and 72 POPC molecules, with a protein:lipid weight ratio circa 2.5:1, suggesting a structure distinct from HDL. High stability of these nanoparticles and their HDL-like size suggest that similar lipoproteins may form in vivo during inflammation or injury when SAA concentration is high and membranes from dead cells require rapid removal. We speculate that solubilization of membranes by SAA to generate lipoproteins in a spontaneous energy-independent process constitutes the primordial function of this ancient protein, providing the first line of defense in clearing cell debris from the injured sites.
Collapse
Affiliation(s)
- Nicholas M Frame
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany St., Boston, MA 02118, USA.
| | - Shobini Jayaraman
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany St., Boston, MA 02118, USA.
| | - Donald L Gantz
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany St., Boston, MA 02118, USA.
| | - Olga Gursky
- Department of Physiology & Biophysics, Boston University School of Medicine, 700 Albany St., Boston, MA 02118, USA.
| |
Collapse
|
84
|
Identification of a panel of serum protein markers in early stage of sepsis and its validation in a cohort of patients. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2017; 51:465-472. [PMID: 28655573 DOI: 10.1016/j.jmii.2016.12.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/18/2016] [Accepted: 12/13/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND Sepsis is a life-threatening illness with a challenging diagnosis. Current serum biomarkers are not sensitive enough for diagnosis. With the aim of finding proteins associated with sepsis, serum protein profile was compared between patients and healthy donors and serum classical inflammatory proteins were analyzed in both groups. METHODS Serum protein profiles were characterized by two-dimensional electrophoresis (2DE). Identification of the proteins was carried out by mass spectrophotometry and their validation was performed by Enzyme-Linked-ImmunoSorbent Assay (ELISA) in a cohort of 85 patients and 67 healthy donors. Seven classical inflammatory proteins were analyzed in the same cohort by ELISA: interleukin-2 receptor α-chain (sCD25), scavenger receptor cysteine-rich-type-1 (sCD163), tumor-necrosis factor receptor superfamily-member-6 (sFas), hemeoxigenase-1 decycling (HO-1), interleukin-6 (IL-6), interleukin-18 (IL-18) and intercellular adhesion-molecule-1 (sICAM-1). RESULTS After 2DE, 20 significantly differently expressed spots were identified by mass spectrometry analysis, revealing deregulation of six different proteins upon sepsis and 50% were validated by ELISA: Antithrombin-III (AT-III), Clusterin (CLUS) and Serum amyloid A-1 (SAA-1). Serum concentration of AT-III and CLUS was significantly lower in patients' serum, whereas SAA-1 showed higher values in septic patients. Serum concentration of the seven inflammatory proteins was significantly increased in septic patients. Functional analysis of the ten deregulated proteins revealed an enrichment of proteins related mainly to the activation of the immune response. CONCLUSION We have identified a panel of ten potential sepsis marker proteins biologically connected and validated in a large number of patients, whose analysis could be considered as a complementary tool for the diagnosis of sepsis.
Collapse
|
85
|
Serum Amyloid A Induces a Vascular Smooth Muscle Cell Phenotype Switch through the p38 MAPK Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2017. [PMID: 28642873 PMCID: PMC5469989 DOI: 10.1155/2017/4941379] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Atherosclerosis is an important pathological condition which is accompanied by a vascular smooth muscle cell (VSMC) phenotype switch toward a synthetic phenotype. As an acute-phase protein, Serum Amyloid A (SAA) is thought to have a close relationship to atherosclerosis development. However, no study has investigated the direct effect of SAA on the VSMC phenotype switch, as well as the underlying mechanisms. The purpose of our study was to explore the effect of SAA on the VSMC phenotype switch and the potential mechanisms involved. In our study, we found that SAA induced the VSMC phenotype switch which reduced expression of the smooth muscle cell (SMC) marker and enhanced expression of the matrix synthesis related marker. The proliferative ability of VSMCs was also increased by SAA treatment. Furthermore, our research found that SAA activated the ERK1/2 and p38 MAPK signaling pathways. Finally, by applying the ERK1/2 and p38 inhibitors, U0126 and SB203580, we demonstrated that the SAA-induced VSMC phenotype switch was p38-dependent. Taken together, these results indicated that SAA may play an important role in promoting the VSMC phenotype switch through the p38 MAPK signaling pathway.
Collapse
|
86
|
Ugurlu S, Hacioglu A, Adibnia Y, Hamuryudan V, Ozdogan H. Tocilizumab in the treatment of twelve cases with aa amyloidosis secondary to familial mediterranean fever. Orphanet J Rare Dis 2017; 12:105. [PMID: 28558744 PMCID: PMC5450086 DOI: 10.1186/s13023-017-0642-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 04/26/2017] [Indexed: 12/11/2022] Open
Abstract
Background There is no established treatment of AA amyloidosis, a long-term complication of various chronic inflammatory diseases associated with increased mortality, such as familial Mediterranian fever (FMF). Recently there are few reports pointing out that tocilizumab(TCZ), an anti IL-6 agent may be effective in AA amyloidosis resistant to conventional treatments. We report our data on the effect of TCZ in patients with FMF complicated with AA amyloidosis. Methods FMF patients with histologically proven AA amyloidosis, treated with TCZ (8 mg/kg per month) were followed monthly and the changes in creatinine, creatinine clearance, the amount of 24-hour urinary protein, erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) were noted throughout the treatment period. Adverse effects of the treatment were closely monitored. Results TCZ was given to 12 patients (6 F, 6 M) who also continued to receive colchicine (1.9 ± 0.4 mg/day). Coexisting diseases were ankylosing spondylitis(4) and Crohn’s disease(1). The mean age was 35.2 ± 10.0 years and the mean follow-up on TCZ was 17.5 ± 14.7 months. The renal functions remained stable (mean creatinine from 1.1 ± 0.9 mg/dl to 1.0 ± 0.6 mg/dl), while a significant decrease in acute phase response (the mean CRP from 18.1 ± 19.5 mg/L to 5.8 ± 7.1 mg/L and ESR from 48.7 ± 31.0 mm/h to 28.7 ± 28.3 mm/h) was observed and the mean 24-hour urinary protein excretion reduced from 6537.6 ± 6526.0 mg/dl to 4745.5 ± 5462.7 mg/dl. Two patients whose renal functions were impaired prior to TCZ therapy improved significantly on this regimen. No infusion reaction was observed. None of the patients experienced any FMF attack under TCZ treatment with the exception of 2, one of whom had less frequent attacks while the other had episodes of erysipelas-like erythema. Conclusıon Tocilizumab improved the acute phase response and the renal function in this group of patients and was generally well tolerated. Besides improving the renal function TCZ seemed to control the recurrence of FMF attacks too. Further studies are warrented to test the efficacy and safety of TCZ in AA amyloidosis secondary to FMF as well as other inflammatory conditions.
Collapse
Affiliation(s)
- Serdal Ugurlu
- Division of Rheumatology, Department of Medicine, Cerrahpasa Medical Faculty, University of Istanbul, Istanbul, Turkey
| | - Aysa Hacioglu
- Division of Rheumatology, Department of Medicine, Cerrahpasa Medical Faculty, University of Istanbul, Istanbul, Turkey
| | - Yasaman Adibnia
- Division of Rheumatology, Department of Medicine, Cerrahpasa Medical Faculty, University of Istanbul, Istanbul, Turkey
| | - Vedat Hamuryudan
- Division of Rheumatology, Department of Medicine, Cerrahpasa Medical Faculty, University of Istanbul, Istanbul, Turkey
| | - Huri Ozdogan
- Division of Rheumatology, Department of Medicine, Cerrahpasa Medical Faculty, University of Istanbul, Istanbul, Turkey.
| |
Collapse
|
87
|
Choi H, Ignacio RMC, Lee ES, Wilson AJ, Khabele D, Son DS. Augmented Serum Amyloid A1/2 Mediated by TNF-induced NF-κB in Human Serous Ovarian Epithelial Tumors. Immune Netw 2017; 17:121-127. [PMID: 28458624 PMCID: PMC5407984 DOI: 10.4110/in.2017.17.2.121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor-α (TNF) is well known to be involved in the immune system and ovarian inflammation. Ovarian cancer is an inflammation-related malignancy that lacks early screening strategies, resulting in late diagnosis followed by high mortality. Based on our previous data, TNF induced abundant serum amyloid A (SAA), an acute phase protein linked to inflammation, in ovarian granulosal cells. To date, the regulation and expression of SAA in ovarian cancer is not fully elucidated. Here, we investigated the relationship between TNF and SAA by comparing human normal ovarian tissues and serous ovarian tumors. We found that SAA1/2 was significantly expressed in tumor tissues, but no or trace expression levels in normal tissues. TNF was also significantly upregulated in ovarian tumor tissues compared to normal tissues. Moreover, TNF significantly increased SAA1/2 levels in human ovarian cancer cell lines, OVCAR-3 and SKOV-3, in a time-dependent manner. Since the SAA1 promoter contains two nuclear factor (NF)-κB sites, we examined whether TNF regulates SAA1 promoter activity. Deletion analysis revealed that the proximal NF-κB site (-95/-85) played a critical role in regulating TNF-induced SAA1 promoter activity. Within 2 h after intraperitoneal injection of lipopolysaccharide, a product known to stimulate release of TNF, SAA preferably localized to ovarian epithelial cells and the thecal-interstitial layers compared to granulosal cell layers. Based on Gene Expression Omnibus (GEO) database, SAA1/2 and TNF were dominantly expressed in advanced grade ovarian cancer. Taken together, the accumulation of SAA1/2 in ovarian cancer could be mediated by TNF-induced NF-κB activation.
Collapse
Affiliation(s)
- Hyeongjwa Choi
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Rosa Mistica C Ignacio
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Eun-Sook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL 32301, USA
| | - Andrew J Wilson
- Department of Obstetrics and Gynecology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Dineo Khabele
- Department of Obstetrics and Gynecology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Deok-Soo Son
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
88
|
Baranova IN, Souza ACP, Bocharov AV, Vishnyakova TG, Hu X, Vaisman BL, Amar MJ, Chen Z, Remaley AT, Patterson AP, Yuen PST, Star RA, Eggerman TL. Human SR-BII mediates SAA uptake and contributes to SAA pro-inflammatory signaling in vitro and in vivo. PLoS One 2017; 12:e0175824. [PMID: 28423002 PMCID: PMC5396919 DOI: 10.1371/journal.pone.0175824] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/01/2017] [Indexed: 12/13/2022] Open
Abstract
Serum amyloid A (SAA) is an acute phase protein with cytokine-like and chemotactic properties, that is markedly up-regulated during various inflammatory conditions. Several receptors, including FPRL-1, TLR2, TLR4, RAGE, class B scavenger receptors, SR-BI and CD36, have been identified as SAA receptors. This study provides new evidence that SR-BII, splice variant of SR-BI, could function as an SAA receptor mediating its uptake and pro-inflammatory signaling. The uptake of Alexa Fluor488 SAA was markedly (~3 fold) increased in hSR-BII-expressing HeLa cells when compared with mock-transfected cells. The levels of SAA-induced interleukin-8 secretion by hSR-BII-expressing HEK293 cells were also significantly (~3-3.5 fold) higher than those detected in control cells. Moderately enhanced levels of phosphorylation of all three mitogen-activated protein kinases, ERK1/2, and p38 and JNK, were observed in hSR-BII-expressing cells following SAA stimulation when compared with control wild type cells. Transgenic mice with pLiv-11-directed liver/kidney overexpression of hSR-BI or hSR-BII were used to assess the in vivo role of each receptor in SAA-induced pro-inflammatory response in these organs. Six hours after intraperitoneal SAA injection both groups of transgenic mice demonstrated markedly higher (~2-5-fold) expression levels of inflammatory mediators in the liver and kidney compared to wild type mice. Histological examinations of hepatic and renal tissue from SAA-treated mice revealed moderate level of damage in the liver of both transgenic but not in the wild type mice. Activities of plasma transaminases, biomarkers of liver injury, were also moderately higher in hSR-B transgenic mice when compared to wild type mice. Our findings identify hSR-BII as a functional SAA receptor that mediates SAA uptake and contributes to its pro-inflammatory signaling via the MAPKs-mediated signaling pathways.
Collapse
Affiliation(s)
- Irina N. Baranova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ana C. P. Souza
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alexander V. Bocharov
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tatyana G. Vishnyakova
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Xuzhen Hu
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Boris L. Vaisman
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Marcelo J. Amar
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Zhigang Chen
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Alan T. Remaley
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amy P. Patterson
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Peter S. T. Yuen
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert A. Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas L. Eggerman
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
89
|
Li W, Wang W, Zuo R, Liu C, Shu Q, Ying H, Sun K. Induction of pro-inflammatory genes by serum amyloid A1 in human amnion fibroblasts. Sci Rep 2017; 7:693. [PMID: 28386088 PMCID: PMC5429602 DOI: 10.1038/s41598-017-00782-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/13/2017] [Indexed: 12/11/2022] Open
Abstract
Serum amyloid A1 (SAA1) is an acute response protein, which is mainly produced by the liver, during infection. However, it remains unknown whether SAA1 can be produced in human fetal membranes where it is able to elicit events pertinent to labor initiation. We demonstrated that SAA1 was expressed in the fibroblasts and epithelium of the amnion and the trophoblasts of the chorion. Further study in human amnion fibroblasts showed that SAA1 production was augmented by interleukin-1β (IL-1β) and cortisol alone and synergistically, and SAA1 in turn induced the expression of IL-1β, interleukin-6 (IL-6), cyclooxygenase-2 (COX-2) and PGE2 production. These effects of SAA1 were mediated through activation of the NF-κB, p38 and ERK1/2 pathways via the toll-like receptor 4 (TLR4). Inhibition of TLR4 attenuated not only SAA1-induced activation of NF-κB, p38 and ERK1/2 but also increases in IL-1β, IL-6 and COX-2 expression. Moreover, SAA1 expression was increased in human amnion tissue following spontaneous labor. In conclusion, this study has demonstrated for the first time that SAA1 can be produced in human fetal membranes, which can be greatly induced in the presence of proinflammatory cytokines and glucocorticoids thereby producing effects associated with parturition.
Collapse
Affiliation(s)
- Wenjiao Li
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, P.R. China
| | - Wangsheng Wang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, P.R. China
| | - Rujuan Zuo
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, P.R. China
| | - Chao Liu
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, P.R. China
| | - Qun Shu
- Department of Obstetrics and Gynecology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Hao Ying
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, P.R. China
| | - Kang Sun
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China.
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, P.R. China.
| |
Collapse
|
90
|
Zatula A, Dikic A, Mulder C, Sharma A, Vågbø CB, Sousa MML, Waage A, Slupphaug G. Proteome alterations associated with transformation of multiple myeloma to secondary plasma cell leukemia. Oncotarget 2017; 8:19427-19442. [PMID: 28038447 PMCID: PMC5386695 DOI: 10.18632/oncotarget.14294] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 11/30/2016] [Indexed: 01/22/2023] Open
Abstract
Plasma cell leukemia is a rare and aggressive plasma cell neoplasm that may either originate de novo (primary PCL) or by leukemic transformation of multiple myeloma (MM) to secondary PCL (sPCL). The prognosis of sPCL is very poor, and currently no standard treatment is available due to lack of prospective clinical studies. In an attempt to elucidate factors contributing to transformation, we have performed super-SILAC quantitative proteome profiling of malignant plasma cells collected from the same patient at both the MM and sPCL stages of the disease. 795 proteins were found to be differentially expressed in the MM and sPCL samples. Gene ontology analysis indicated a metabolic shift towards aerobic glycolysis in sPCL as well as marked down-regulation of enzymes involved in glycan synthesis, potentially mediating altered glycosylation of surface receptors. There was no significant change in overall genomic 5-methylcytosine or 5-hydroxymethylcytosine at the two stages, indicating that epigenetic dysregulation was not a major driver of transformation to sPCL. The present study constitutes the first attempt to provide a comprehensive map of the altered protein expression profile accompanying transformation of MM to sPCL in a single patient, identifying several candidate proteins that can be targeted by currently available small molecule drugs. Our dataset furthermore constitutes a reference dataset for further proteomic analysis of sPCL transformation.
Collapse
Affiliation(s)
- Alexey Zatula
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Aida Dikic
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Celine Mulder
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Present address: University of Utrecht, Utrecht, Holland
| | - Animesh Sharma
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Cathrine B Vågbø
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Mirta M L Sousa
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Anders Waage
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Department of Hematology, Department of Medicine, St. Olav's Hospital, Trondheim, Norway
| | - Geir Slupphaug
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,PROMEC Core Facility for Proteomics and Metabolomics, Norwegian University of Science and Technology, NTNU, Trondheim, and the Central Norway Regional Health Authority, Stjørdal, Norway
| |
Collapse
|
91
|
Agostini-Vulaj D, Sharma AK, Findeis-Hosey JJ, McMahon LA, Gonzalez RS. Distinction between inflammatory hepatocellular adenoma and mass effect on liver sampling. Hum Pathol 2017; 61:105-110. [DOI: 10.1016/j.humpath.2016.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 11/18/2016] [Accepted: 12/01/2016] [Indexed: 10/20/2022]
|
92
|
Linke RP, Meinel A, Chalcroft JP, Urieli-Shoval S. Serum amyloid A (SAA) treatment enhances the recovery of aggravated polymicrobial sepsis in mice, whereas blocking SAA's invariant peptide results in early death. Amyloid 2017; 24:149-150. [PMID: 28434296 DOI: 10.1080/13506129.2017.1295950] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
| | - Angelika Meinel
- a Reference Center of Amyloid Diseases , Martinsried , Germany
| | - James P Chalcroft
- b Max Planck Institute of Neurobiology , Martinsried , Germany , and
| | | |
Collapse
|
93
|
Ibrahim MI, Ramy AR, Abdelhamid AS, Ellaithy MI, Omar A, Harara RM, Fathy H, Abolouz AS. Maternal serum amyloid A level as a novel marker of primary unexplained recurrent early pregnancy loss. Int J Gynaecol Obstet 2017; 136:298-303. [PMID: 28099717 DOI: 10.1002/ijgo.12076] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 10/25/2016] [Accepted: 12/05/2016] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To assess maternal serum amyloid A (SAA) levels among women with primary unexplained recurrent early pregnancy loss (REPL). METHODS A prospective study was conducted among women with missed spontaneous abortion in the first trimester at Ain Shams University Maternity Hospital, Cairo, Egypt, between January 21 and December 25, 2014. Women with at least two consecutive primary unexplained REPLs and no previous live births were enrolled. A control group was formed of women with no history of REPL who had at least one previous uneventful pregnancy with no adverse outcomes. Serum samples were collected to measure SAA levels. The main outcome was the association between SAA and primary unexplained REPL. RESULTS Each group contained 96 participants. Median SAA level was significantly higher among women with REPL (50.0 μg/mL, interquartile range 26.0-69.0) than among women in the control group (11.6 μg/mL, interquartile range 6.2-15.5; P<0.001). The SAA level was an independent indicator of primary unexplained REPL, after adjusting for maternal age and gestational age (odds ratio 1.12, 95% confidence interval 1.06-1.19; P<0.001). CONCLUSION Elevated SAA levels found among women with primary unexplained REPL could represent a novel biomarker for this complication of pregnancy.
Collapse
Affiliation(s)
- Moustafa I Ibrahim
- Department of Obstetrics and Gynecology, Ain Shams University Faculty of Medicine, Cairo, Egypt
| | - Ahmed R Ramy
- Department of Obstetrics and Gynecology, Ain Shams University Faculty of Medicine, Cairo, Egypt
| | - Ahmed S Abdelhamid
- Department of Obstetrics and Gynecology, Ain Shams University Faculty of Medicine, Cairo, Egypt
| | - Mohamed I Ellaithy
- Department of Obstetrics and Gynecology, Ain Shams University Faculty of Medicine, Cairo, Egypt
| | - Amna Omar
- Department of Obstetrics and Gynecology, Ain Shams University Faculty of Medicine, Cairo, Egypt
| | - Rany M Harara
- Department of Obstetrics and Gynecology, Ain Shams University Faculty of Medicine, Cairo, Egypt
| | - Hayam Fathy
- Department of Obstetrics and Gynecology, Ain Shams University Faculty of Medicine, Cairo, Egypt
| | - Ashraf S Abolouz
- Department of Obstetrics and Gynecology, October 6th University Faculty of Medicine, Cairo, Egypt
| |
Collapse
|
94
|
Proteins behaving badly. Substoichiometric molecular control and amplification of the initiation and nature of amyloid fibril formation: lessons from and for blood clotting. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 123:16-41. [DOI: 10.1016/j.pbiomolbio.2016.08.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 08/14/2016] [Accepted: 08/19/2016] [Indexed: 02/08/2023]
|
95
|
Gaspar BL, Garg C, Vasishta RK, Nada R, Goyal MK. Localised SAA amyloidosis with intra-axonal intra-myelin amyloid deposits. Pathology 2016; 49:103-105. [PMID: 27913045 DOI: 10.1016/j.pathol.2016.08.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/16/2016] [Accepted: 08/24/2016] [Indexed: 11/27/2022]
Affiliation(s)
| | | | | | | | - Manoj Kumar Goyal
- Department of Neurology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
96
|
El-Deeb WM, Elmoslemany AM. The diagnostic accuracy of acute phase proteins and proinflammatory cytokines in sheep with pneumonic pasteurellosis. PeerJ 2016; 4:e2161. [PMID: 27547520 PMCID: PMC4957991 DOI: 10.7717/peerj.2161] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/01/2016] [Indexed: 11/20/2022] Open
Abstract
The goal of this study was to assess the diagnostic accuracy of acute phase proteins and proinflammatory cytokines in sheep with pneumonic pasteurellosis. Blood samples were collected from 56 sheep (36 naturally infected with Pasteurella multocida and 20 healthy controls) belonging to one farm in Eastern region, Saudi Arabia. Serum samples were evaluated for acute phase proteins (Haptoglobin (Hp), serum amyloid A (SAA) and fibrinogen (Fb)), and the proinflammatory cytokines (interleukins (IL-1α, IL-1β, and IL-6), tumor necrosis factor-alpha (TNF-α), and interferon-gamma (IFN-ϒ)). Additionally, nasopharyngeal swabs and bronchoalveolar lavages were collected from all animals for bacteriological examinations. Receiver operating characteristic curve was used to assess the diagnostic performance of each parameter. All parameters showed moderate to high degree of positive correlation with case-control status. There was no significant difference in the area under the curve (AUC) among acute phase proteins; however, both Hp and SAA showed better sensitivity and specificity than Fb. The proinflammatory cytokines (IL1-α, IL1-β, and IL6) showed similar and highly accurate diagnostic performance (AUC > 0.9), whereas IFN-ϒ was moderately accurate (AUC = 0.79). In conclusion, this study confirms the value of acute phase proteins and cytokines as diagnostic biomarkers of naturally occuring pneumonic pasteurellosis in sheep.
Collapse
Affiliation(s)
- Wael M El-Deeb
- Department of Clinical Studies, College of Veterinary Medicine and Animal Resources, King Faisal University, Al-Ahsa, Saudi Arabia; Department of Internal Medicine, Infectious Diseases and Fish Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed M Elmoslemany
- Department of Clinical Studies, College of Veterinary Medicine and Animal Resources, King Faisal University, Al-Ahsa, Saudi Arabia; Faculty of Veterinary Medicine, Hygiene and Preventive Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| |
Collapse
|
97
|
Perez JD, Sakata MM, Colucci JA, Spinelli GA, Felipe CR, Carvalho VM, Cardozo KHM, Medina-Pestana JO, Tedesco-Silva H, Schor N, Casarini DE. Plasma proteomics for the assessment of acute renal transplant rejection. Life Sci 2016; 158:111-20. [PMID: 27393492 DOI: 10.1016/j.lfs.2016.06.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/08/2016] [Accepted: 06/28/2016] [Indexed: 12/13/2022]
Abstract
UNLABELLED Renal transplant is the best treatment for patients with chronical kidney disease however acute graft rejection is the major impediment to success in renal transplantation leading to loss of the organ the first year after transplantation. The aim of this study was to identify plasma proteins that may be early biomarkers of acute rejection of renal allograft, developing a diagnostic model that avoids the loss of the transplanted organ. Shotgun proteomics (LC-MS/MS) method was used to analyze a set of thirty-one plasma samples, including 06 from patients with acute graft rejection after transplantation (rejection group/Rej-group) and twenty-five from renal transplant patients with stable renal graft function (control group/Ct-group). As results nineteen proteins were upregulated in the rejection group compared to the control group, and two proteins were downregulated; and three were present exclusively in the rejection group. After analysis, we selected four proteins that were related to the acute phase response and that were strongly associated with each other: they are alpha-1 antitrypsin (A1AT), alpha-2 antiplasmin (A2AP), serum amyloid A (SAA) and apolipoprotein CIII (APOC3). We think that simultaneous monitoring of SAA and APOC3 can provide insights into a broad profile of signaling proteins and is highly valuable for the early detection of a possible acute renal graft rejection. STATEMENT OF SIGNIFICANCE OF THE STUDY In this study we did plasma shotgun patients with and without acute rejection of renal allograft. In a clinical setting an acute rejection is typically suspected upon an increase in plasma creatinine and renal biopsy. But these methods are late and unspecific; sometimes the rejection process is already advanced when there is an increase in serum creatinine. Therefore, it is necessary to find proteins that can predict the allograft rejection process. In our study were able to identify changes in the concentration of plasma protein belonging to a network of protein interaction processes the acute phase response. We believe, therefore, that development of a routine diagnosis of these proteins can detect early acute rejection of renal allograft process, thus preventing its loss.
Collapse
Affiliation(s)
- Juliana D Perez
- Department of Medicine, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maísa M Sakata
- Department of Medicine, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Juliana A Colucci
- Universidade de Santo Amaro, Programa em Medicina Populacional, São Paulo, Brazil
| | - Gláucio A Spinelli
- Nephrology Division, Hospital do Rim e Hipertensão, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Claudia R Felipe
- Nephrology Division, Hospital do Rim e Hipertensão, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | - José O Medina-Pestana
- Nephrology Division, Hospital do Rim e Hipertensão, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Hélio Tedesco-Silva
- Nephrology Division, Hospital do Rim e Hipertensão, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Nestor Schor
- Department of Medicine, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Dulce E Casarini
- Department of Medicine, Division of Nephrology, Universidade Federal de São Paulo, São Paulo, Brazil.
| |
Collapse
|
98
|
Connolly M, Rooney PR, McGarry T, Maratha AX, McCormick J, Miggin SM, Veale DJ, Fearon U. Acute serum amyloid A is an endogenous TLR2 ligand that mediates inflammatory and angiogenic mechanisms. Ann Rheum Dis 2016; 75:1392-8. [PMID: 26290589 DOI: 10.1136/annrheumdis-2015-207655] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 08/01/2015] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Acute-phase serum amyloid A (A-SAA) has cytokine-like properties and is expressed at sites of inflammation. We examined whether A-SAA-induced pro-inflammatory mechanisms are mediated through Toll-like receptor 2 (TLR2) in rheumatoid arthritis (RA). METHODS The effect of A-SAA on human embryonic kidney (HEK), TLR2 or TLR4 cells was quantified by nuclear factor (NF)-κB luciferase reporter assays. A-SAA-induced RASFC and dHMVEC function were performed in the presence of a specific neutralising anti-TLR2 mAb (OPN301) (1 μg/mL) and matched IgG isotype control Ab (1 μg/mL). Cell surface expression of intracellular adhesion molecule (ICAM)-1, chemokine expression, cell migration, invasion and angiogenesis were assessed by flow cytometry, ELISA, Matrigel invasion chambers and tube formation assays. MyD88 expression was assessed by real-time PCR and western blot. RESULTS A-SAA induced TLR2 activation through induction of NF-κB (p<0.05), but failed to induce NF-κB in HEK-TLR4 cells, confirming specificity for TLR2. A-SAA-induced proliferation, invasion and migration were significantly inhibited in the presence of anti-TLR2 (all p<0.05), with no significant effect observed for tumour necrosis factor-α-induced events. Additionally, A-SAA-induced ICAM-1, interleukin-8, monocyte chemoattractant protein-1, RANTES and GRO-α expression were significantly reduced in the presence of anti-TLR2 (all p<0.05), as was A-SAA induced angiogenesis (p<0.05). Finally, A-SAA induced MyD88 signalling in RASFC and dHMVEC (p<0.05). CONCLUSIONS A-SAA is an endogenous ligand for TLR2, inducing pro-inflammatory effects in RA. Blocking the A-SAA/TLR2 interaction may be a potential therapeutic intervention in RA.
Collapse
Affiliation(s)
- Mary Connolly
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| | - Peter R Rooney
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| | - Trudy McGarry
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| | - Ashwini X Maratha
- Department of Biology, Institute of Immunology, National University of Ireland Maynooth, Maynooth, County Kildare, Ireland
| | - Jennifer McCormick
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| | - Sinead M Miggin
- Department of Biology, Institute of Immunology, National University of Ireland Maynooth, Maynooth, County Kildare, Ireland
| | - Douglas J Veale
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| | - Ursula Fearon
- Centre for Arthritis and Rheumatic Diseases, Dublin Academic Medical Centre and Conway Institute of Biomolecular and Biomedical Research, Dublin 4, Ireland
| |
Collapse
|
99
|
Benard EL, Rougeot J, Racz PI, Spaink HP, Meijer AH. Transcriptomic Approaches in the Zebrafish Model for Tuberculosis-Insights Into Host- and Pathogen-specific Determinants of the Innate Immune Response. ADVANCES IN GENETICS 2016; 95:217-51. [PMID: 27503359 DOI: 10.1016/bs.adgen.2016.04.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Mycobacterium marinum infection in zebrafish has become a well-established model of tuberculosis. Both embryonic and adult zebrafish infection studies have contributed to our knowledge of the development and function of tuberculous granulomas, which are typical of mycobacterial pathogenesis. In this review we discuss how transcriptome profiling studies have helped to characterize this infection process. We illustrate this using new RNA sequencing (RNA-Seq) data that reveals three main phases in the host response to M. marinum during the early stages of granuloma development in zebrafish embryos and larvae. The early phase shows induction of complement and transcription factors, followed by a relatively minor induction of pro-inflammatory cytokines within hours following phagocytosis of M. marinum. A minimal response is observed in the mid-phase, between 6 hours and 1day post infection, when the tissue dissemination of M. marinum begins. During subsequent larval development the granulomas expand and a late-phase response is apparent, which is characterized by progressively increasing induction of complement, transcription factors, pro-inflammatory cytokines, matrix metalloproteinases, and other defense and inflammation-related gene groups. This late-phase response shares common components with the strong and acute host transcriptome response that has previously been reported for Salmonella typhimurium infection in zebrafish embryos. In contrast, the early/mid-phase response to M. marinum infection, characterized by suppressed pro-inflammatory signaling, is strikingly different from the acute response to S. typhimurium infection. Furthermore, M. marinum infection shows a collective and strongly fluctuating regulation of lipoproteins, while S. typhimurium infection has pronounced effects on amino acid metabolism and glycolysis.
Collapse
Affiliation(s)
- E L Benard
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | - J Rougeot
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | - P I Racz
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | - H P Spaink
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | - A H Meijer
- Institute of Biology, Leiden University, Leiden, The Netherlands
| |
Collapse
|
100
|
Ortega-Martínez I, Gardeazabal J, Erramuzpe A, Sanchez-Diez A, Cortés J, García-Vázquez MD, Pérez-Yarza G, Izu R, Luís Díaz-Ramón J, de la Fuente IM, Asumendi A, Boyano MD. Vitronectin and dermcidin serum levels predict the metastatic progression of AJCC I-II early-stage melanoma. Int J Cancer 2016; 139:1598-607. [PMID: 27216146 PMCID: PMC5089559 DOI: 10.1002/ijc.30202] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 05/11/2016] [Indexed: 01/03/2023]
Abstract
Like many cancers, an early diagnosis of melanoma is fundamental to ensure a good prognosis, although an important proportion of stage I-II patients may still develop metastasis during follow-up. The aim of this work was to discover serum biomarkers in patients diagnosed with primary melanoma that identify those at a high risk of developing metastasis during the follow-up period. Proteomic and mass spectrophotometry analysis was performed on serum obtained from patients who developed metastasis during the first years after surgery for primary tumors and compared with that from patients who remained disease-free for more than 10 years after surgery. Five proteins were selected for validation as prognostic factors in 348 melanoma patients and 100 controls by ELISA: serum amyloid A and clusterin; immune system proteins; the cell adhesion molecules plakoglobin and vitronectin and the antimicrobial protein dermcidin. Compared to healthy controls, melanoma patients have high serum levels of these proteins at the moment of melanoma diagnosis, although the specific values were not related to the histopathological stage of the tumors. However, an analysis based on classification together with multivariate statistics showed that tumor stage, vitronectin and dermcidin levels were associated with the metastatic progression of patients with early-stage melanoma. Although melanoma patients have increased serum dermcidin levels, the REPTree classifier showed that levels of dermcidin <2.98 μg/ml predict metastasis in AJCC stage II patients. These data suggest that vitronectin and dermcidin are potent biomarkers of prognosis, which may help to improve the personalized medical care of melanoma patients and their survival.
Collapse
Affiliation(s)
- Idoia Ortega-Martínez
- Department of Cell Biology and Histology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Jesús Gardeazabal
- Department of Dermatology, Ophthalmology and Otorhinolaryngology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,BioCruces Health Research Institute, Plaza De Cruces S/N, Barakaldo, Bizkaia, Spain
| | - Asier Erramuzpe
- BioCruces Health Research Institute, Plaza De Cruces S/N, Barakaldo, Bizkaia, Spain
| | - Ana Sanchez-Diez
- Department of Dermatology, Ophthalmology and Otorhinolaryngology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,BioCruces Health Research Institute, Plaza De Cruces S/N, Barakaldo, Bizkaia, Spain
| | - Jesús Cortés
- Department of Cell Biology and Histology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,BioCruces Health Research Institute, Plaza De Cruces S/N, Barakaldo, Bizkaia, Spain.,Ikerbasque: The Basque Foundation for Science, Bilbao, Spain
| | - María D García-Vázquez
- Department of Cell Biology and Histology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Gorka Pérez-Yarza
- Department of Cell Biology and Histology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,BioCruces Health Research Institute, Plaza De Cruces S/N, Barakaldo, Bizkaia, Spain
| | - Rosa Izu
- Department of Dermatology, Ophthalmology and Otorhinolaryngology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,BioCruces Health Research Institute, Plaza De Cruces S/N, Barakaldo, Bizkaia, Spain
| | - Jose Luís Díaz-Ramón
- Department of Dermatology, Ophthalmology and Otorhinolaryngology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,BioCruces Health Research Institute, Plaza De Cruces S/N, Barakaldo, Bizkaia, Spain
| | - Ildefonso M de la Fuente
- Institute of Parasitology and Biomedicine Lopez-Neyra, Parque Tecnológico Ciencias De La Salud, Avenida Del Conocimiento S/N, Armilla, Granada, Spain
| | - Aintzane Asumendi
- Department of Cell Biology and Histology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,BioCruces Health Research Institute, Plaza De Cruces S/N, Barakaldo, Bizkaia, Spain
| | - María D Boyano
- Department of Cell Biology and Histology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.,BioCruces Health Research Institute, Plaza De Cruces S/N, Barakaldo, Bizkaia, Spain
| |
Collapse
|