1
|
Katina N, Marchenkov V, Lapteva Y, Balobanov V, Ilyina N, Ryabova N, Evdokimov S, Suvorina M, Surin A, Glukhov A. Authentic hSAA related with AA amyloidosis: New method of purification, folding and amyloid polymorphism. Biophys Chem 2024; 313:107293. [PMID: 39004034 DOI: 10.1016/j.bpc.2024.107293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/04/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
The secondary amyloidosis of humans is caused by the formation of hSAA fibrils in different organs and tissues. Until now hSAA was thought to have low amyloidogenicity in vitro and the majority of SAA aggregation experiments were done using murine protein or hSAA non-pathogenic isoforms. In this work a novel purification method for recombinant hSAA was introduced, enabling to obtain monomeric protein capable of amyloid aggregation under physiological conditions. The stability and amyloid aggregation of hSAA have been examined using a wide range of biophysical methods. It was shown that the unfolding of monomeric protein occurs through the formation of molten globule-like intermediate state. Polymorphism of hSAA amyloids was discovered to depend on the solution pH. At pH 8.5, rapid protein aggregation occurs, which leads to the formation of twisted short fibrils. Even a slight decrease of the pH to 7.8 results in delayed aggregation with the formation of long straight amyloids composed of laterally associated protofilaments. Limited proteolysis experiments have shown that full-length hSAA is involved in the formation of intermolecular interactions in both amyloid polymorphs. The results obtained, and the experimental approach used in this study can serve as a basis for further research on the mechanism of authentic hSAA amyloid formation.
Collapse
Affiliation(s)
- Natalya Katina
- Branch of the Institute of Bioorganic Chemistry RAS, Prospekt Nauki, 6, Pushchino, 142290, Russia; Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| | - Victor Marchenkov
- Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| | - Yulia Lapteva
- Institute for Biological Instrumentation RAS, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Nauki av., 3, Pushchino, 142290, Russia.
| | - Vitalii Balobanov
- Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| | - Nelly Ilyina
- Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| | - Natalya Ryabova
- Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| | | | - Mariya Suvorina
- Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| | - Alexey Surin
- Branch of the Institute of Bioorganic Chemistry RAS, Prospekt Nauki, 6, Pushchino, 142290, Russia; Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia; State Research Center for Applied Microbiology and Biotechnology, Kvartal A, 24, Obolensk, 142279, Russia.
| | - Anatoly Glukhov
- Institute of Protein Research RAS, Institutskaya, 4, Pushchino, 142290, Russia.
| |
Collapse
|
2
|
Abstract
COVID-19 infections decrease total cholesterol, LDL-C, HDL-C, and apolipoprotein A-I, A-II, and B levels while triglyceride levels may be increased or inappropriately normal for the poor nutritional status. The degree of reduction in total cholesterol, LDL-C, HDL-C, and apolipoprotein A-I are predictive of mortality. With recovery lipid/lipoprotein levels return towards pre-infection levels and studies have even suggested an increased risk of dyslipidemia post-COVID-19 infection. The potential mechanisms for these changes in lipid and lipoprotein levels are discussed. Decreased HDL-C and apolipoprotein A-I levels measured many years prior to COVID-19 infections are associated with an increased risk of severe COVID-19 infections while LDL-C, apolipoprotein B, Lp (a), and triglyceride levels were not consistently associated with an increased risk. Finally, data suggest that omega-3-fatty acids and PCSK9 inhibitors may reduce the severity of COVID-19 infections. Thus, COVID-19 infections alter lipid/lipoprotein levels and HDL-C levels may affect the risk of developing COVID-19 infections.
Collapse
|
3
|
Chiriac IE, Vilceanu N, Maghiar A, Andrei C, Hanganu B, Daina LG, Dunarintu S, Buhas LC. Maternal Serum Amyloid A as a Marker of Preterm Birth/PROM: A Systematic Review and Meta-Analysis. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1025. [PMID: 37374228 DOI: 10.3390/medicina59061025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/02/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023]
Abstract
Background and Objectives: Preterm birth, one of the leading causes of neonatal mortality, occurs in between 5 and 18% of births. Premature birth can be induced by a variety of triggers, including infection or inflammation. Serum amyloid A, a family of apolipoproteins, increases significantly and rapidly at the onset of inflammation. This study aims to systematically review the results of studies in the literature that have examined the correlation between SAA and PTB/PROM. Materials and Methods: To examine the correlation between serum amyloid A levels in women who gave birth prematurely, a systematic analysis was performed according to PRISMA guidelines. Studies were retrieved by searching the electronic databases PubMed and Google Scholar. The primary outcome measure was the standardized mean difference in serum amyloid A level comparing the preterm birth or premature rupture of membranes groups and the term birth group. Results: Based on the inclusion criteria, a total of 5 manuscripts adequately addressed the desired outcome and were thus included in the analysis. All included studies showed a statistically significant difference in serum SAA levels between the preterm birth or preterm rupture of membranes groups and the term birth group. The pooled effect, according to the random effects model, is SMD = 2.70. However, the effect is not significant (p = 0.097). In addition, the analysis reveals an increased heterogeneity with an I2 = 96%. Further, the analysis of the influence on heterogeneity found a study that has a significant influence on heterogeneity. However, even after outline exclusion, heterogeneity remained high I2 = 90.7%. Conclusions: There is an association between increased levels of SAA and preterm birth/PROM, but studies have shown great heterogeneity.
Collapse
Affiliation(s)
- Ioana-Evelina Chiriac
- Doctoral School, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Narcis Vilceanu
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Adrian Maghiar
- Department of Surgical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Csep Andrei
- Department of Psycho-Neuroscience and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Bianca Hanganu
- Legal-Medicine Department, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy of Iasi, 700115 Iasi, Romania
| | - Lucia Georgeta Daina
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Simona Dunarintu
- Department of Radiology and Medical Imaging, Timisoara County Emergency Clinical Hospital, 300723 Timisoara, Romania
| | - Liana-Camelia Buhas
- Department of Morphological Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
4
|
Karam S, Haidous M, Royal V, Leung N. Renal AA amyloidosis: presentation, diagnosis, and current therapeutic options: a review. Kidney Int 2023; 103:473-484. [PMID: 36502873 DOI: 10.1016/j.kint.2022.10.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 12/13/2022]
Abstract
Amyloid A amyloidosis is thought to be the second most common form of systemic amyloidosis behind amyloidosis secondary to monoclonal Ig. It is the result of deposition of insoluble fibrils in the extracellular space of tissues and organs derived from the precursor protein serum amyloid A, an acute phase reactant synthesized excessively in the setting of chronic inflammation. The kidney is the most frequent organ involved. Most patients present with proteinuria and kidney failure. The diagnosis is made through tissue biopsy with involvement of the glomeruli in most cases, but also often of the vessels and the tubulointerstitial compartment. The treatment usually targets the underlying etiology and consists increasingly of blocking the inflammatory cascade of cytokines with interleukin-1 inhibitors, interleukin-6 inhibitors, and tumor necrosis factor-α inhibitors to reduce serum amyloid A protein formation. This strategy has also shown efficacy in cases where an underlying etiology cannot be readily identified and has significantly improved the prognosis of this entity. In addition, there has been increased interest at developing effective therapies able to clear amyloid deposits from tissues, albeit with mitigated results so far.
Collapse
Affiliation(s)
- Sabine Karam
- Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, Minnesota, USA.
| | - Mohamad Haidous
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Virginie Royal
- Division of Pathology, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Quebec, Canada
| | - Nelson Leung
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA; Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
5
|
Jiang Q, Sherlock DN, Elolimy AA, Vailati-Riboni M, Yoon I, Loor JJ. Impact of a Saccharomyces cerevisiae fermentation product during an intestinal barrier challenge in lactating Holstein cows on ileal microbiota and markers of tissue structure and immunity. J Anim Sci 2023; 101:skad309. [PMID: 37721866 PMCID: PMC10630188 DOI: 10.1093/jas/skad309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/16/2023] [Indexed: 09/20/2023] Open
Abstract
Feeding a Saccharomyces cerevisiae fermentation product (SCFP; NutriTek, Diamond V, Cedar Rapids, IA) during periods of metabolic stress is beneficial to the health of dairy cows partially through its effect on the gut microbiota. Whether SCFP alters the ileal microbiota in lactating cows during intestinal challenges induced by feed restriction (FR) is not known. We used 16S rRNA sequencing to assess if feeding SCFP during FR to induce gut barrier dysfunction alters microbiota profiles in the ileum. The mRNA abundance of key genes associated with tissue structures and immunity was also detected. Multiparous cows (97.1 ± 7.6 days in milk (DIM); n = 7 per treatment) fed a control diet or the control plus 19 g/d NutriTek for 9 wk were subjected to an FR challenge for 5 d, during which they were fed 40% of their ad libitum intake from the 7 d before FR. All cows were slaughtered at the end of FR. DNA extracted from ileal digesta was subjected to PacBio Full-Length 16S rRNA gene sequencing. High-quality amplicon sequence analyses were performed with Targeted Amplicon Diversity Analysis and MicrobiomeAnalyst. Functional analysis was performed and analyzed using PICRUSt and STAMP. Feeding SCFP did not (P > 0.05) alter dry matter intake, milk yield, or milk components during FR. In addition, SCFP supplementation tended (P = 0.07) to increase the relative abundance of Proteobacteria and Bifidobacterium animalis. Compared with controls, feeding SCFP increased the relative abundance of Lactobacillales (P = 0.03). Gluconokinase, oligosaccharide reducing-end xylanase, and 3-hydroxy acid dehydrogenase were among the enzymes overrepresented (P < 0.05) in response to feeding SCFP. Cows fed SCFP had a lower representation of adenosylcobalamin biosynthesis I (early cobalt insertion) and pyrimidine deoxyribonucleotides de novo biosynthesis III (P < 0.05). Subsets of the Firmicutes genus, Bacteroidota phylum, and Treponema genus were correlated with the mRNA abundance of genes associated with ileal integrity (GCNT3, GALNT5, B3GNT3, FN1, ITGA2, LAMB2) and inflammation (AOX1, GPX8, CXCL12, CXCL14, CCL4, SAA3). Our data indicated that the moderate FR induced dysfunction of the ileal microbiome, but feeding SCFP increased the abundance of some beneficial gut probiotic bacteria and other species related to tissue structures and immunity.
Collapse
Affiliation(s)
- Qianming Jiang
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Danielle N Sherlock
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Ahmed A Elolimy
- Animal Production Department, National Research Center, Dokki, Giza 12622, Egypt
| | | | | | - Juan J Loor
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
6
|
Martínez-Ayala P, Alanis-Sánchez GA, Álvarez-Zavala M, Sánchez-Reyes K, Ruiz-Herrera VV, Cabrera-Silva RI, González-Hernández LA, Ramos-Becerra C, Cardona-Muñoz E, Andrade-Villanueva JF. Effect of antiretroviral therapy on decreasing arterial stiffness, metabolic profile, vascular and systemic inflammatory cytokines in treatment-naïve HIV: A one-year prospective study. PLoS One 2023; 18:e0282728. [PMID: 36930649 PMCID: PMC10022802 DOI: 10.1371/journal.pone.0282728] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/21/2023] [Indexed: 03/18/2023] Open
Abstract
INTRODUCTION Cardiovascular disease is a major cause of death among people living with HIV (PLH). Non-treated PLH show increased levels of inflammation and biomarkers of vascular activation, and arterial stiffness as a prognostic cardiovascular disease risk factor. We investigated the effect of one year of ART on treatment-naïve HIV(+) individuals on arterial stiffness and inflammatory and vascular cytokines. METHODS We cross-sectionally compared aortic stiffness via tonometry, inflammatory, and vascular serum cytokines on treatment-naïve (n = 20) and HIV (-) (n = 9) matched by age, sex, metabolic profile, and Framingham score. We subsequently followed young, treatment-naïve individuals after 1-year of ART and compared aortic stiffness, metabolic profile, and inflammatory and vascular serum biomarkers to baseline. Inflammatory biomarkers included: hs-CRP, D-Dimer, SAA, sCD163s, MCP-1, IL-8, IL-18, MRP8/14. Vascular cytokines included: myoglobin, NGAL, MPO, Cystatin C, ICAM-1, VCAM-1, and MMP9. RESULTS Treatment-naïve individuals were 34.8 years old, mostly males (95%), and with high smoking prevalence (70%). Baseline T CD4+ was 512±324 cells/mcL. cfPWV was similar between HIV(-) and treatment-naïve (6.8 vs 7.3 m/s; p = 0.16) but significantly decreased after ART (-0.52 m/s; 95% CI -0.87 to -0.16; p0.006). Almost all the determined cytokines were significantly higher compared to controls, except for MCP-1, myoglobin, NGAL, cystatin C, and MMP-9. At follow-up, only total cholesterol and triglycerides increased and all inflammatory cytokines significantly decreased. Regarding vascular cytokines, MPO, ICAM-1, and VCAM-1 showed a reduction. D-Dimer tended to decrease (p = 0.06) and hs-CRP did not show a significant reduction (p = 0.17). CONCLUSION One year of ART had a positive effect on reducing inflammatory and vascular cytokines and arterial stiffness.
Collapse
Affiliation(s)
- Pedro Martínez-Ayala
- HIV Unit Department, University Hospital "Fray Antonio Alcalde", University of Guadalajara, Guadalajara, Mexico
| | | | - Monserrat Álvarez-Zavala
- Clinical Medicine Department, HIV and Immunodeficiencies Research Institute, CUCS-University of Guadalajara, Guadalajara, Mexico
| | - Karina Sánchez-Reyes
- Clinical Medicine Department, HIV and Immunodeficiencies Research Institute, CUCS-University of Guadalajara, Guadalajara, Mexico
| | - Vida Verónica Ruiz-Herrera
- HIV Unit Department, University Hospital "Fray Antonio Alcalde", University of Guadalajara, Guadalajara, Mexico
| | - Rodolfo Ismael Cabrera-Silva
- Clinical Medicine Department, HIV and Immunodeficiencies Research Institute, CUCS-University of Guadalajara, Guadalajara, Mexico
| | - Luz Alicia González-Hernández
- HIV Unit Department, University Hospital "Fray Antonio Alcalde", University of Guadalajara, Guadalajara, Mexico
- Clinical Medicine Department, HIV and Immunodeficiencies Research Institute, CUCS-University of Guadalajara, Guadalajara, Mexico
| | - Carlos Ramos-Becerra
- Department of Physiology, Arterial Stiffness Laboratory, University of Guadalajara, Guadalajara, Mexico
| | - Ernesto Cardona-Muñoz
- Department of Physiology, Arterial Stiffness Laboratory, University of Guadalajara, Guadalajara, Mexico
| | - Jaime Federico Andrade-Villanueva
- HIV Unit Department, University Hospital "Fray Antonio Alcalde", University of Guadalajara, Guadalajara, Mexico
- Clinical Medicine Department, HIV and Immunodeficiencies Research Institute, CUCS-University of Guadalajara, Guadalajara, Mexico
- * E-mail:
| |
Collapse
|
7
|
Sil S, Thangaraj A, Oladapo A, Hu G, Kutchy NA, Liao K, Buch S, Periyasamy P. Role of Autophagy in HIV-1 and Drug Abuse-Mediated Neuroinflammaging. Viruses 2022; 15:44. [PMID: 36680084 PMCID: PMC9866731 DOI: 10.3390/v15010044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Chronic low-grade inflammation remains an essential feature of HIV-1 infection under combined antiretroviral therapy (cART) and contributes to the accelerated cognitive defects and aging in HIV-1 infected populations, indicating cART limitations in suppressing viremia. Interestingly, ~50% of the HIV-1 infected population on cART that develops cognitive defects is complicated by drug abuse, involving the activation of cells in the central nervous system (CNS) and neurotoxin release, altogether leading to neuroinflammation. Neuroinflammation is the hallmark feature of many neurodegenerative disorders, including HIV-1-associated neurocognitive disorders (HAND). Impaired autophagy has been identified as one of the underlying mechanisms of HAND in treated HIV-1-infected people that also abuse drugs. Several lines of evidence suggest that autophagy regulates CNS cells' responses and maintains cellular hemostasis. The impairment of autophagy is associated with low-grade chronic inflammation and immune senescence, a known characteristic of pathological aging. Therefore, autophagy impairment due to CNS cells, such as neurons, microglia, astrocytes, and pericytes exposure to HIV-1/HIV-1 proteins, cART, and drug abuse could have combined toxicity, resulting in increased neuroinflammation, which ultimately leads to accelerated aging, referred to as neuroinflammaging. In this review, we focus on the potential role of autophagy in the mechanism of neuroinflammaging in the context of HIV-1 and drug abuse.
Collapse
Affiliation(s)
- Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Annadurai Thangaraj
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Centre for Excellence in Nanobio Translational Research, Anna University, BIT Campus, Tiruchirappalli 620024, Tamil Nadu, India
| | - Abiola Oladapo
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Naseer A Kutchy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Ke Liao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA 90048, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
8
|
Brynge M, Gardner R, Sjöqvist H, Karlsson H, Dalman C. Maternal levels of acute phase proteins in early pregnancy and risk of autism spectrum disorders in offspring. Transl Psychiatry 2022; 12:148. [PMID: 35393396 PMCID: PMC8989993 DOI: 10.1038/s41398-022-01907-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 01/08/2023] Open
Abstract
Previous research supports a contribution of early-life immune disturbances in the etiology of autism spectrum disorders (ASD). Biomarker studies of the maternal innate (non-adaptive) immune status related to ASD risk have focused on one of the acute phase proteins (APP), C-reactive protein (CRP), with conflicting results. We evaluated levels of eight different APP in first-trimester maternal serum samples, from 318 mothers to ASD cases and 429 mothers to ASD-unaffected controls, nested within the register-based Stockholm Youth Cohort. While no overall associations between high levels of APP and ASD were observed, associations varied across diagnostic sub-groups based on co-occurring conditions. Maternal levels of CRP in the lowest compared to the middle tertile were associated with increased risk of ASD without ID or ADHD in offspring (OR = 1.92, 95% CI 1.08-3.42). Further, levels of maternal ferritin in the lowest (OR = 1.78, 95% CI 1.18-2.69) and highest (OR = 1.64, 95% CI 1.11-2.43) tertiles were associated with increased risk of any ASD diagnosis in offspring, with stronger associations still between the lowest (OR = 3.81, 95% CI 1.91-7.58) and highest (OR = 3.36, 95% CI 1.73-6.53) tertiles of ferritin and risk of ASD with ID. The biological interpretation of lower CRP levels among mothers to ASD cases is not clear but might be related to the function of the maternal innate immune system. The finding of aberrant levels of ferritin conferring risk of ASD-phenotypes indicates a plausibly important role of iron during neurodevelopment.
Collapse
Affiliation(s)
- Martin Brynge
- Department of Global Public Health, Karolinska Institutet, Stockholm, 17177, Sweden.
| | - Renee Gardner
- Department of Global Public Health, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Hugo Sjöqvist
- Department of Global Public Health, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Håkan Karlsson
- Department of Neuroscience, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Christina Dalman
- Department of Global Public Health, Karolinska Institutet, Stockholm, 17177, Sweden
- Centre for Epidemiology and Community Medicine, Region Stockholm, Stockholm, 17129, Sweden
| |
Collapse
|
9
|
Pomorska-Mól M, Libera K, Larska M, Krzysiak MK. Acute-phase protein concentrations in serum of clinically healthy and diseased European bison (Bison bonasus) - preliminary study. BMC Vet Res 2022; 18:28. [PMID: 35012560 PMCID: PMC8744219 DOI: 10.1186/s12917-021-03133-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This is the first report describing levels of APPs in European bison. Serum concentration of acute phase proteins (APPs) may be helpful to assess general health status in wildlife and potentially useful in selecting animals for elimination. Since there is a lack of literature data regarding concentration of APPs in European bisons, establishment of the reference values is also needed. METHODS A total of 87 European bison from Polish populations were divided into two groups: (1) healthy: immobilized for transportation, placing a telemetry collar and routine diagnostic purposes; and (2) selectively culled due to the poor health condition. The serum concentration of haptoglobin, serum amyloid A and α1-acid-glycoprotein were determined using commercial quantitative ELISA assays. Since none of the variables met the normality assumptions, non-parametric Mann-Whitney U test was used for all comparisons. Statistical significance was set at p < 0.05. Statistical analyses were performed using Statistica 13.3 (Tibco, USA). RESULTS The concentration of haptoglobin and serum amyloid A was significantly higher in animals culled (euthanised) due to the poor condition in respect to the clinically healthy European bison. The levels of α1-acid-glycoprotein did not show statistical difference between healthy and sick animals. CONCLUSIONS Correlation between APPs concertation and health status was proven, therefore the determination of selected APPs may be considered in future as auxiliary predictive tool in assessing European bison health condition.
Collapse
Affiliation(s)
- Małgorzata Pomorska-Mól
- Department of Preclinical Sciences and Infectious Diseases, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, ul. Wołyńska 35, 60-637, Poznań, Poland.
| | - Kacper Libera
- Department of Preclinical Sciences and Infectious Diseases, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, ul. Wołyńska 35, 60-637, Poznań, Poland
| | - Magdalena Larska
- Department of Virology, National Veterinary Research Institute, Al. Partyzantów 57, 24-100, Puławy, Poland
| | - Michał K Krzysiak
- Białowieża National Park, Park Pałacowy 11, 17-230, Białowieża, Poland.,Institute of Forest Sciences, Faculty of Civil Engineering and Environmental Sciences, Bialystok University of Technology, Białystok, Poland
| |
Collapse
|
10
|
Uncomplicated Plasmodium vivax malaria: mapping the proteome from circulating platelets. Clin Proteomics 2022; 19:1. [PMID: 34991449 PMCID: PMC8903537 DOI: 10.1186/s12014-021-09337-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 12/21/2021] [Indexed: 01/07/2023] Open
Abstract
Background Thrombocytopenia is frequent in Plasmodium vivax malaria but the role of platelets in pathogenesis is unknown. Our study explores the platelet (PLT) proteome from uncomplicated P. vivax patients, to fingerprint molecular pathways related to platelet function. Plasma levels of Platelet factor 4 (PF4/CXCL4) and Von Willebrand factor (VWf), as well as in vitro PLTs—P. vivax infected erythrocytes (Pv-IEs) interactions were also evaluated to explore the PLT response and effect on parasite development. Methods A cohort of 48 patients and 25 healthy controls were enrolled. PLTs were purified from 5 patients and 5 healthy controls for Liquid Chromatography–Mass spectrometry (LC–MS/MS) analysis. Plasma levels of PF4/CXCL4 and VWf were measured in all participants. Additionally, P. vivax isolates (n = 10) were co-cultured with PLTs to measure PLT activation by PF4/CXCL4 and Pv-IE schizonts formation by light microscopy. Results The proteome from uncomplicated P. vivax patients showed 26 out of 215 proteins significantly decreased. PF4/CXCL4 was significantly decreased followed by other proteins involved in platelet activation, cytoskeletal remodeling, and endothelial adhesion, including glycoprotein V that was significantly decreased in thrombocytopenic patients. In contrast, acute phase proteins, including SERPINs and Amyloid Serum A1 were increased. High levels of VWf in plasma from patients suggested endothelial activation while PF4/CXCL4 plasma levels were similar between patients and controls. Interestingly, high levels of PF4/CXCL4 were released from PLTs—Pv-IEs co-cultures while Pv-IEs schizont formation was inhibited. Conclusions The PLT proteome analyzed in this study suggests that PLTs actively respond to P. vivax infection. Altogether, our findings suggest important roles of PF4/CXCL4 during uncomplicated P. vivax infection through a possible intracellular localization. Our study shows that platelets are active responders to P. vivax infection, inhibiting intraerythrocytic parasite development. Future studies are needed to further investigate the molecular pathways of interaction between platelet proteins found in this study and host response, which could affect parasite control as well as disease progression. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-021-09337-7.
Collapse
|
11
|
Potential Novel Biomarkers for Mastitis Diagnosis in Sheep. Animals (Basel) 2021; 11:ani11102783. [PMID: 34679803 PMCID: PMC8532728 DOI: 10.3390/ani11102783] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Inflammation of the mammary gland (mastitis) is an important disease of dairy sheep. Mastitis management depends mainly on the diagnosis. Conventional diagnostic methods including somatic cell count, California Mastitis Test, and microbial culture have limitations. Therefore researchers are looking for new diagnostic biomarkers of mastitis including specific proteins produced by the liver in case of disease (acute phase proteins), unique genetic sequences (miRNAs), or antimicrobial peptides produced by immune cells during inflammation (cathelicidines). Abstract This review aims to characterize promising novel markers of ovine mastitis. Mastitis is considered as one of the primary factors for premature culling in dairy sheep and has noticeable financial, productional, and animal welfare-related implications. Furthermore, clinical, and subclinical mammary infections negatively affect milk yield and alter the milk composition, thereby leading to lowered quality of dairy products. It is, therefore, crucial to control and prevent mastitis through proper diagnosis, treatment or culling, and appropriate udder health management particularly at the end of the lactation period. The clinical form of mastitis is characterized by abnormalities in milk and mammary gland tissue alteration or systemic symptoms consequently causing minor diagnostic difficulties. However, to identify ewes with subclinical mastitis, laboratory diagnostics is crucial. Mastitis control is primarily dependent on determining somatic cell count (SCC) and the California Mastitis Test (CMT), which aim to detect the quantity of cells in the milk sample. The other useful diagnostic tool is microbial culture, which complements SCC and CMT. However, all mentioned diagnostic methods have their limitations and therefore novel biomarkers of ovine subclinical mastitis are highly desired. These sensitive indicators include acute-phase proteins, miRNA, and cathelicidins measurements, which could be determined in ovine serum and/or milk and in the future may become useful in early mastitis diagnostics as well as a preventive tool. This may contribute to increased detection of ovine mammary gland inflammation in sheep, especially in subclinical form, and consequently improves milk quality and quantity.
Collapse
|
12
|
Yang L, Liu S, Han S, Hu Y, Wu Z, Shi X, Pang B, Ma Y, Jin J. The HDL from septic-ARDS patients with composition changes exacerbates pulmonary endothelial dysfunction and acute lung injury induced by cecal ligation and puncture (CLP) in mice. Respir Res 2020; 21:293. [PMID: 33148285 PMCID: PMC7640393 DOI: 10.1186/s12931-020-01553-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 10/20/2020] [Indexed: 01/13/2023] Open
Abstract
Background Septic-acute respiratory distress syndrome (ARDS), characterized by the acute lung injury (ALI) secondary to aberrant systemic inflammatory response, has high morbidity and mortality. Despite increased understanding of ALI pathogenesis, the therapies to prevent lung dysfunction underlying systemic inflammatory disorder remain elusive. The high density lipoprotein (HDL) has critical protective effects in sepsis and its dysfunction has a manifested contribution to septic organ failure. However, the adverse changes in HDL composition and function in septic-ARDS patients are large unknown. Methods To investigate HDL remodeling in septic-ARDS, we analyzed the changes of HDL composition from 40 patients with septic-ARDS (A-HDL) and 40 matched normal controls (N-HDL). To determine the deleterious functional remodeling of HDL, A-HDL or N-HDL was administrated to C57BL/6 and apoA-I knock-out (KO) mice after cecal ligation and puncture (CLP) procedure. Mouse lung microvascular endothelial cells (MLECs) were further treated by these HDLs to investigate whether the adverse effects of A-HDL were associated with endothelial dysfunction. Results Septic-ARDS patients showed significant changes of HDL composition, accompanied with significantly decreased HDL-C. We further indicated that A-HDL treatment aggravated CLP induced ALI. Intriguingly, these deleterious effects of A-HDL were associated with pulmonary endothelial dysfunction, rather than the increased plasma lipopolysaccharide (LPS). Further in vitro results demonstrated the direct effects of A-HDL on MLECs, including increased endothelial permeability, enhanced expressions of adhesion proteins and pro-inflammatory cytokines via activating NF-κB signaling and decreased junction protein expression. Conclusions Our results depicted the remodeling of HDL composition in sepsis, which predisposes lung to ARDS via inducing ECs dysfunction. These results also demonstrated the importance of circulating HDL in regulating alveolar homeostasis.
Collapse
Affiliation(s)
- Liu Yang
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan road, Beijing Chaoyang Hospital Jingxi Branch, Beijing, China.,Beijing Institute of Respiratory Medicine, Beijing, China
| | - Sijie Liu
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan road, Beijing Chaoyang Hospital Jingxi Branch, Beijing, China
| | - Silu Han
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan road, Beijing Chaoyang Hospital Jingxi Branch, Beijing, China
| | - Yuhan Hu
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan road, Beijing Chaoyang Hospital Jingxi Branch, Beijing, China
| | - Zhipeng Wu
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan road, Beijing Chaoyang Hospital Jingxi Branch, Beijing, China
| | - Xiaoqian Shi
- The Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Baosen Pang
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan road, Beijing Chaoyang Hospital Jingxi Branch, Beijing, China.,Beijing Institute of Respiratory Medicine, Beijing, China.,The Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yingmin Ma
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan road, Beijing Chaoyang Hospital Jingxi Branch, Beijing, China. .,Beijing Institute of Respiratory Medicine, Beijing, China.
| | - Jiawei Jin
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan road, Beijing Chaoyang Hospital Jingxi Branch, Beijing, China. .,Beijing Institute of Respiratory Medicine, Beijing, China. .,The Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
13
|
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: 33] [Impact Index Per Article: 6.6] [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
|
14
|
Ibrahim MI, Ellaithy MI, Hussein AM, Nematallah MM, Allam HA, Abdelhamid AS, Harara RM, Riad AA, Rafaat TA. Measurement of maternal serum amyloid A as a novel marker of preterm birth. J Matern Fetal Neonatal Med 2019; 34:2467-2472. [PMID: 31522581 DOI: 10.1080/14767058.2019.1668370] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To study the association between maternal serum amyloid A (mSAA) levels and preterm birth (PTB). METHODS This prospective observational nested case control study was conducted at Ain Shams University Maternity Hospital, Cairo, Egypt, between May 2017 and December 2017. The study recruited pregnant women at 26-34 weeks presented with threatened preterm labor (PTL). Women with PTB were included in cases group while control group included women who continued pregnancy and delivered at term. Serum samples were collected to measure mSAA levels. The main outcome of the study was the association between mSAA levels and PTB. Secondary outcomes included neonatal intensive care unit admissions and neonatal mortality. RESULTS Fifty-eight women were included in the final analysis (29 in each group). Women with PTB had a statistically significant higher mSAA levels [5.1 (4.5-7.7) vs. 1.2 (0.0-2.5) mg/l, for cases and controls respectively, p < .001]. Higher mSAA levels were also observed among women whose babies were admitted to NICU, but there was no significant relation between mSAA level and neonatal death. A statistically significant negative correlation was found between mSAA and gestational age at delivery and neonatal birth weight. mSAA had an excellent value to predict PTB (AUC = 0.972 [95% CI, 0.891-0.998], p < .0001), fair value to predict admission to NICU and a poor value to predict neonatal death. CONCLUSIONS mSAA level was found to be elevated among women with threatened PTL who end with PTB; mSAA is a potentially useful predictive marker of PTB that warrant further study. CLINICALTRIALS.GOV: NCT01639027.
Collapse
Affiliation(s)
- Moustafa I Ibrahim
- Obstetrics and Gynecology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed I Ellaithy
- Obstetrics and Gynecology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed M Hussein
- Obstetrics and Gynecology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mona M Nematallah
- Obstetrics and Gynecology Department, Kafr Eldawar Hospital, Elbeheira, Egypt
| | - Heba A Allam
- Obstetrics and Gynecology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed S Abdelhamid
- Obstetrics and Gynecology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Rany M Harara
- Obstetrics and Gynecology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Amr A Riad
- Obstetrics and Gynecology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Tarek A Rafaat
- Obstetrics and Gynecology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
15
|
Vieyres G, Pietschmann T. HCV Pit Stop at the Lipid Droplet: Refuel Lipids and Put on a Lipoprotein Coat before Exit. Cells 2019; 8:cells8030233. [PMID: 30871009 PMCID: PMC6468556 DOI: 10.3390/cells8030233] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 02/07/2023] Open
Abstract
The replication cycle of the liver-tropic hepatitis C virus (HCV) is tightly connected to the host lipid metabolism, during the virus entry, replication, assembly and egress stages, but also while the virus circulates in the bloodstream. This interplay coins viral particle properties, governs viral cell tropism, and facilitates immune evasion. This review summarizes our knowledge of these interactions focusing on the late steps of the virus replication cycle. It builds on our understanding of the cell biology of lipid droplets and the biosynthesis of liver lipoproteins and attempts to explain how HCV hijacks these organelles and pathways to assemble its lipo-viro-particles. In particular, this review describes (i) the mechanisms of viral protein translocation to and from the lipid droplet surface and the orchestration of an interface between replication and assembly complexes, (ii) the importance of the triglyceride mobilization from the lipid droplets for HCV assembly, (iii) the interplay between HCV and the lipoprotein synthesis pathway including the role played by apolipoproteins in virion assembly, and finally (iv) the consequences of these complex virus–host interactions on the virion composition and its biophysical properties. The wealth of data accumulated in the past years on the role of the lipid metabolism in HCV assembly and its imprint on the virion properties will guide vaccine design efforts and reinforce our understanding of the hepatic lipid metabolism in health and disease.
Collapse
Affiliation(s)
- Gabrielle Vieyres
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), 30625 Hannover, Germany.
| | - Thomas Pietschmann
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), 30625 Hannover, Germany.
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany.
| |
Collapse
|
16
|
Page MJ, Thomson GJA, Nunes JM, Engelbrecht AM, Nell TA, de Villiers WJS, de Beer MC, Engelbrecht L, Kell DB, Pretorius E. Serum amyloid A binds to fibrin(ogen), promoting fibrin amyloid formation. Sci Rep 2019; 9:3102. [PMID: 30816210 PMCID: PMC6395759 DOI: 10.1038/s41598-019-39056-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/16/2019] [Indexed: 12/15/2022] Open
Abstract
Complex associations exist between inflammation and thrombosis, with the inflammatory state tending to promote coagulation. Fibrinogen, an acute phase protein, has been shown to interact with the amyloidogenic ß-amyloid protein of Alzheimer’s disease. However, little is known about the association between fibrinogen and serum amyloid A (SAA), a highly fibrillogenic protein that is one of the most dramatically changing acute phase reactants in the circulation. To study the role of SAA in coagulation and thrombosis, in vitro experiments were performed where purified human SAA, in concentrations resembling a modest acute phase response, was added to platelet-poor plasma (PPP) and whole blood (WB), as well as purified and fluorescently labelled fibrinogen. Results from thromboelastography (TEG) suggest that SAA causes atypical coagulation with a fibrin(ogen)-mediated increase in coagulation, but a decreased platelet/fibrin(ogen) interaction. In WB scanning electron microscopy analysis, SAA mediated red blood cell (RBC) agglutination, platelet activation and clumping, but not platelet spreading. Following clot formation in PPP, the presence of SAA increased amyloid formation of fibrin(ogen) as determined both with auto-fluorescence and with fluorogenic amyloid markers, under confocal microcopy. SAA also binds to fibrinogen, as determined with a fluorescent-labelled SAA antibody and correlative light electron microscopy (CLEM). The data presented here indicate that SAA can affect coagulation by inducing amyloid formation in fibrin(ogen), as well as by propelling platelets to a more prothrombotic state. The discovery of these multiple and complex effects of SAA on coagulation invite further mechanistic analyses.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa
| | - Greig J A Thomson
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa
| | - J Massimo Nunes
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa
| | - Theo A Nell
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa
| | - Willem J S de Villiers
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa.,Department of Internal Medicine, Stellenbosch University, Stellenbosch, South Africa
| | - Maria C de Beer
- Department of Physiology, Saha Cardiovascular Research Center and Barnstable Brown Diabetes Center, University of Kentucky, Lexington, KY, USA
| | - Lize Engelbrecht
- Fluorescence Microscopy Unit, Central Analytical Facilities, Stellenbosch University, Stellenbosch, South Africa
| | - Douglas B Kell
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa. .,School of Chemistry, The University of Manchester, 131 Princess St, MANCHESTER M1 7DN, Lancs, UK. .,The Manchester Institute of Biotechnology, The University of Manchester, 131 Princess St, MANCHESTER M1 7DN, Lancs, UK. .,Department of Biochemistry, Institute of integrative Biology, Biosciences Building., University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1 Matieland, 7602, Stellenbosch, South Africa.
| |
Collapse
|
17
|
Di Iorio B, Di Micco L, Bruzzese D, Nardone L, Russo L, Formisano P, D'Esposito V, Russo D. Ultrapure dialysis water obtained with additional ultrafilter may reduce inflammation in patients on hemodialysis. J Nephrol 2017; 30:795-801. [PMID: 28840494 PMCID: PMC5698401 DOI: 10.1007/s40620-017-0422-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 07/11/2017] [Indexed: 11/24/2022]
Abstract
BACKGROUND Patients on standard dialysis, in particular those on high-flux and high-efficiency dialysis, are exposed to hundreds of liters of dialysis-water per week. The quality of dialysis-water is a factor responsible for inflammation in dialysis patients. Inflammation is a potent trigger of atherosclerosis and a pathogenetic factor in anemia, increasing mortality and morbidity in dialysis patients. Current systems for water treatment do not completely eliminate bacteria and endotoxins. This prospective study tested whether improved dialysis-water purity by an additional ultrafilter can reduce inflammation and ameliorate hemoglobin levels, with a consequent reduction in erythropoietin-stimulating agents (ESA). METHODS An ultrafilter, composed of two serially positioned devices with polysulfone membranes of 2.0 and 1.0 m2, respectively, was positioned within the fluid pathway before the dialysis machine. Prevalent dialysis patients were assigned either to continue dialysis with conventional dialysis-water (control phase) or to initiate dialysis sessions with improved dialysis-water purity (study phase). After 6 months, patients were crossed over. Total study duration was 1 year. Routine chemistry, bacterial count, endotoxin levels in dialysis-water as well as blood levels of pro- and anti-inflammatory cytokines, human serum amyloid A, C-reactive protein and fraction 5 of complement were measured. RESULTS Thirty-two patients completed the study. Mean bacterial count was lower and endotoxin levels were absent in dialysis-water obtained with the ultrafilter. At the end of the study-phase, C-reactive protein and pro-inflammatory cytokines decreased while anti-inflammatory ones increased. Hemoglobin levels were improved with lower ESA doses. CONCLUSIONS An additional ultrafilter improved dialysis-water purity, reduced levels of inflammation markers, ameliorated hemoglobin concentration with reduced ESA doses. These results remain speculative but they may generate studies to assess whether improved dialysis-water quality with an ultrafilter can reduce inflammation and improve survival of dialysis patients.
Collapse
Affiliation(s)
- Biagio Di Iorio
- Department of Nephrology, "A. Landolfi" Hospital (Solofra, Avellino), Solofra, Italy
| | - Lucia Di Micco
- Department of Nephrology, "A. Landolfi" Hospital (Solofra, Avellino), Solofra, Italy
| | - Dario Bruzzese
- Department of Statistics, University of Naples "FEDERICO II", Naples, Italy
| | - Luca Nardone
- Department of Nephrology, "A. Landolfi" Hospital (Solofra, Avellino), Solofra, Italy
| | - Luigi Russo
- Department of Public Health, University of Naples "FEDERICO II", Naples, Italy
| | - Pietro Formisano
- Department of Genetics, University of Naples "FEDERICO II", Naples, Italy
| | | | - Domenico Russo
- Department of Public Health, University of Naples "FEDERICO II", Naples, Italy.
| |
Collapse
|
18
|
Iqbal F, Baker WS, Khan MI, Thukuntla S, McKinney KH, Abate N, Tuvdendorj D. Current and future therapies for addressing the effects of inflammation on HDL cholesterol metabolism. Br J Pharmacol 2017; 174:3986-4006. [PMID: 28326542 PMCID: PMC5660004 DOI: 10.1111/bph.13743] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/16/2017] [Accepted: 02/02/2017] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Inflammatory processes arising from metabolic abnormalities are known to precipitate the development of CVD. Several metabolic and inflammatory markers have been proposed for predicting the progression of CVD, including high density lipoprotein cholesterol (HDL-C). For ~50 years, HDL-C has been considered as the atheroprotective 'good' cholesterol because of its strong inverse association with the progression of CVD. Thus, interventions to increase the concentration of HDL-C have been successfully tested in animals; however, clinical trials were unable to confirm the cardiovascular benefits of pharmaceutical interventions aimed at increasing HDL-C levels. Based on these data, the significance of HDL-C in the prevention of CVD has been called into question. Fundamental in vitro and animal studies suggest that HDL-C functionality, rather than HDL-C concentration, is important for the CVD-preventive qualities of HDL-C. Our current review of the literature positively demonstrates the negative impact of systemic and tissue (i.e. adipose tissue) inflammation in the healthy metabolism and function of HDL-C. Our survey indicates that HDL-C may be a good marker of adipose tissue health, independently of its atheroprotective associations. We summarize the current findings on the use of anti-inflammatory drugs to either prevent HDL-C clearance or improve the function and production of HDL-C particles. It is evident that the therapeutic agents currently available may not provide the optimal strategy for altering HDL-C metabolism and function, and thus, further research is required to supplement this mechanistic approach for preventing the progression of CVD. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Fatima Iqbal
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Wendy S Baker
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Madiha I Khan
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Shwetha Thukuntla
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Kevin H McKinney
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Nicola Abate
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| | - Demidmaa Tuvdendorj
- Division of Endocrinology, Department of Internal MedicineUniversity of Texas Medical BranchGalvestonTXUSA
| |
Collapse
|
19
|
Feng Z, Long W, Hao B, Ding D, Ma X, Zhao L, Pang X. A human stool-derived Bilophila wadsworthia strain caused systemic inflammation in specific-pathogen-free mice. Gut Pathog 2017; 9:59. [PMID: 29090023 PMCID: PMC5657053 DOI: 10.1186/s13099-017-0208-7] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/17/2017] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Bilophila wadsworthia is a major member of sulfidogenic bacteria in human gut, it was originally recovered from different clinical specimens of intra-abdominal infections and recently was reported potentially linked to different chronic metabolic disorders. However, there is still insufficient understanding on its detailed function and mechanism to date. METHODS A B. wadsworthia strain was isolated from fresh feces of a latent autoimmune diabetes in adults patient and we investigated its pathogenicity by oral administration to specific-pathogen-free mice. Tissue samples and serum were collected after sacrifice. Stool samples were collected at different time points to profile the gut microbiota. RESULTS Bilophila wadsworthia infection resulted in the reduction of body weight and fat mass, apparent hepatosplenomegaly and elevated serum inflammatory factors, including serum amyloid A and interleukin-6, while without significant change of the overall gut microbiota structure. CONCLUSIONS These results demonstrated that higher amount of B. wadsworthia caused systemic inflammatory response in SPF mice, which adds new evidence to the pathogenicity of this bacterium and implied its potential role to the chronic inflammation related metabolic diseases like diabetes.
Collapse
Affiliation(s)
- Zhou Feng
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai, 200240 China
| | - Wenmin Long
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai, 200240 China
| | - Binhan Hao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai, 200240 China
| | - Ding Ding
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai, 200240 China
| | - Xiaoqing Ma
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai, 200240 China
| | - Liping Zhao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai, 200240 China
| | - Xiaoyan Pang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai, 200240 China
| |
Collapse
|
20
|
Claus S, Puscalau-Girtu I, Walther P, Syrovets T, Simmet T, Haupt C, Fändrich M. Cell-to-cell transfer of SAA1 protein in a cell culture model of systemic AA amyloidosis. Sci Rep 2017; 7:45683. [PMID: 28361953 PMCID: PMC5374501 DOI: 10.1038/srep45683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/02/2017] [Indexed: 12/15/2022] Open
Abstract
Systemic AA amyloidosis arises from the misfolding of serum amyloid A1 (SAA1) protein and the deposition of AA amyloid fibrils at multiple sites within the body. Previous research already established that mononuclear phagocytes are crucial for the formation of the deposits in vivo and exposure of cultures of such cells to SAA1 protein induces the formation of amyloid deposits within the culture dish. In this study we show that both non-fibrillar and fibrillar SAA1 protein can be readily transferred between cultured J774A.1 cells, a widely used model of mononuclear phagocytes. We find that the exchange is generally faster with non-fibrillar SAA1 protein than with fibrils. Exchange is blocked if cells are separated by a membrane, while increasing the volume of cell culture medium had only small effects on the observed exchange efficiency. Taken together with scanning electron microscopy showing the presence of the respective types of physical interactions between the cultured cells, we conclude that the transfer of SAA1 protein depends on direct cell-to-cell contacts or tunneling nanotubes.
Collapse
Affiliation(s)
- Stephanie Claus
- Institute of Protein Biochemistry, Ulm University, Helmholtzstr. 8/1, 89081 Ulm, Germany
| | - Ioana Puscalau-Girtu
- Institute of Protein Biochemistry, Ulm University, Helmholtzstr. 8/1, 89081 Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, Albert-Einstein-Allee 11, 89069 Ulm, Germany
| | - Tatiana Syrovets
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Helmholtzstr. 20, 89081 Ulm, Germany
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Helmholtzstr. 20, 89081 Ulm, Germany
| | - Christian Haupt
- Institute of Protein Biochemistry, Ulm University, Helmholtzstr. 8/1, 89081 Ulm, Germany
| | - Marcus Fändrich
- Institute of Protein Biochemistry, Ulm University, Helmholtzstr. 8/1, 89081 Ulm, Germany
| |
Collapse
|
21
|
Jayaraman S, Haupt C, Gursky O. Paradoxical effects of SAA on lipoprotein oxidation suggest a new antioxidant function for SAA. J Lipid Res 2016; 57:2138-2149. [PMID: 27744369 DOI: 10.1194/jlr.m071191] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/12/2016] [Indexed: 01/01/2023] Open
Abstract
Oxidative stress and inflammation, which involve a dramatic increase in serum amyloid A (SAA) levels, are critical in the development of atherosclerosis. Most SAA circulates on plasma HDL particles, altering their cardioprotective properties. SAA-enriched HDL has diminished anti-oxidant effects on LDL, which may contribute to atherogenesis. We determined combined effects of SAA enrichment and oxidation on biochemical changes in HDL. Normal human HDLs were incubated with SAA, oxidized by various factors (Cu2+, myeloperoxidase, H2O2, OCl-), and analyzed for lipid and protein modifications and biophysical remodeling. Three novel findings are reported: addition of SAA reduces oxidation of HDL and LDL lipids; oxidation of SAA-containing HDL in the presence of OCl- generates a covalent heterodimer of SAA and apoA-I that resists the release from HDL; and mild oxidation promotes spontaneous release of proteins (SAA and apoA-I) from SAA-enriched HDL. We show that the anti-oxidant effects of SAA extend to various oxidants and are mediated mainly by the unbound protein. We propose that free SAA sequesters lipid hydroperoxides and delays lipoprotein oxidation, though much less efficiently than other anti-oxidant proteins, such as apoA-I, that SAA displaces from HDL. These findings prompt us to reconsider the role of SAA in lipid oxidation in vivo.
Collapse
Affiliation(s)
- Shobini Jayaraman
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118
| | - Christian Haupt
- Institute of Protein Biochemistry, University of Ulm, 89081 Ulm, Germany
| | - Olga Gursky
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118
| |
Collapse
|
22
|
Jang WY, Lee BR, Jeong J, Sung Y, Choi M, Song P, Kim H, Jang S, Kim H, Joo KI, Lee JW, Choo YS, Kim E, Ryoo ZY. Overexpression of serum amyloid a 1 induces depressive-like behavior in mice. Brain Res 2016; 1654:55-65. [PMID: 27608955 DOI: 10.1016/j.brainres.2016.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 08/30/2016] [Accepted: 09/04/2016] [Indexed: 11/27/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by loss of memory and cognitive abilities. In AD, amyloid β (Aβ) protein aggregates in the brain of patients, forming amyloid plaques. Aβ plaques are known to be surrounded by activated microglial cells. Serum amyloid A (SAA) is elevated from several hundred to 1000-fold as part of the immune response against various injuries, including trauma, infection, and inflammation. Additionally, continuous elevation of SAA is related to the development of amyloidosis. This study was designed to identify the relationship between SAA1 and AD using liver specific SAA1 overexpressing mice (TG), because SAA1 is expressed in the liver during the acute phase. We detected exogenous SAA1 expression in the brain of TG mice. This result implies that liver-derived SAA1 migrates to the brain tissues. Thus, we confirmed that the blood brain barrier (BBB) functioned normally using Evans-blue staining and CARS. Furthermore, our results show an increase in the accumulation of the 87kDa form of Aβ in TG mice compared to wild type mice (WT). Additionally, the number of microglial cells and levels of pro-inflammatory cytokines were increased. Next, we investigated the relationship between SAA1 and depression by performing social interaction tests. The results showed that TG mice have a tendency to avoid stranger mice and an impaired social recognition. In conclusion, the SAA1 TG mouse model is a valuable model to study depression.
Collapse
Affiliation(s)
- Woo Young Jang
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Bo-Ram Lee
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea; Division of Nano & Energy Convergence Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Republic of Korea
| | - Jain Jeong
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Younghun Sung
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Minjee Choi
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Park Song
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Hyerim Kim
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Soyoung Jang
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Hyunmin Kim
- Division of Nano & Energy Convergence Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Republic of Korea
| | - Kyung-Il Joo
- Division of Nano & Energy Convergence Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Republic of Korea
| | - Jeong-Woong Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Yeon Sik Choo
- Department of Biology, Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Eunjoo Kim
- Division of Nano & Energy Convergence Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Republic of Korea.
| | - Zae Young Ryoo
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea.
| |
Collapse
|
23
|
Aspichueta P, Pérez-Agote B, Pérez S, Ochoa B, Fresnedo O. Impaired response of VLDL lipid and apoB secretion to endotoxin in the fasted rat liver. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519060120030501] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Bacterial infection elicits hypertriglyceridemia attributed to increased hepatic production of very low-density lipoprotein (VLDL) particles and decreased peripheral metabolism. The mechanisms underlying VLDL overproduction in sepsis are as yet unclear, but seem to be fed/fasted state-dependent. To learn more about this, we investigated hepatocytes isolated from fasted rats, made endotoxic by 1 mg/kg lipopolysaccharide (LPS) injection, for their ability to secrete the VLDL protein and lipid components. The results were then related to lipogenesis markers and expression of genes critical to VLDL biogenesis. Endotoxic rats showed increased levels of serum VLDL-apoB (10-fold), -triglyceride (2-fold), and -cholesterol (2-fold), whereby circulating VLDL were lipid-poor particles. Similarly, VLDL-apoB secretion by isolated endotoxic hepatocytes was ~85% above control, whereas marginal changes in the output of VLDL-lipid classes occurred. This was accompanied by a substantial rise in apoB and a moderate rise in MTP mRNA levels, but with basal de novo formation and efficiency of secretion of triglycerides, cholesterol and cholesteryl esters. These results indicate that during periods of food restriction, endotoxin does not enhance lipid provision to accomplish normal lipidation of overproduced apoB molecules, though this does occur to a sufficient extent to pass the proteasome checkpoint and secretion of lipid-poor, type 2 VLDL takes place.
Collapse
Affiliation(s)
- Patricia Aspichueta
- Department of Physiology, University of the Basque Country Medical School, Bilbao, Spain
| | - Begoña Pérez-Agote
- Department of Physiology, University of the Basque Country Medical School, Bilbao, Spain
| | - Silvia Pérez
- Department of Physiology, University of the Basque Country Medical School, Bilbao, Spain
| | - Begoña Ochoa
- Department of Physiology, University of the Basque Country Medical School, Bilbao, Spain,
| | - Olatz Fresnedo
- Department of Physiology, University of the Basque Country Medical School, Bilbao, Spain
| |
Collapse
|
24
|
Trenchevska O, Yassine HN, Borges CR, Nelson RW, Nedelkov D. Development of quantitative mass spectrometric immunoassay for serum amyloid A. Biomarkers 2016; 21:743-751. [PMID: 27308834 DOI: 10.1080/1354750x.2016.1201533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Proteins can exist as multiple proteoforms in vivo that can have important roles in physiological and pathological states. METHODS We present the development and characterization of mass spectrometric immunoassay (MSIA) for quantitative determination of serum amyloid A (SAA) proteoforms. RESULTS Intra- and inter-day precision revealed CVs <10%. Against existing SAA ELISA, the developed MSIA showed good correlation according to the Altman-Bland plot. Individual concentrations of the SAA proteoforms across a cohort of 170 samples revealed 7 diverse SAA polymorphic types and 12 different proteoforms. CONCLUSION The new SAA MSIA enables parallel analysis of SAA polymorphisms and quantification of all expressed SAA proteoforms, in a high-throughput and time-efficient manner.
Collapse
Affiliation(s)
| | - Hussein N Yassine
- b Department of Medicine , University of Southern California , Los Angeles , CA , USA
| | - Chad R Borges
- a The Biodesign Institute, Arizona State University , Tempe , AZ , USA
| | - Randall W Nelson
- a The Biodesign Institute, Arizona State University , Tempe , AZ , USA
| | - Dobrin Nedelkov
- a The Biodesign Institute, Arizona State University , Tempe , AZ , USA
| |
Collapse
|
25
|
Rosani U, Domeneghetti S, Gerdol M, Franzoi M, Pallavicini A, Venier P. Serum amyloid A in marine bivalves: An acute phase and innate immunity protein. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 59:136-144. [PMID: 26828389 DOI: 10.1016/j.dci.2016.01.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/26/2016] [Accepted: 01/27/2016] [Indexed: 06/05/2023]
Abstract
Serum amyloid A (SAA) is among the most potent acute phase proteins (APP) in vertebrates. After injury, its early expression can dramatically increase to promote the recruitment of immuno-competent cells, expression of pro-inflammatory proteins and the activation of the innate immune defences. Although APP have been studied in many vertebrates, only recently their search was extended to invertebrates and the finding of SAA-like molecules has opened new questions on the immune-regulatory functions of these soluble proteins in the animal kingdom. Taking advantage of the considerable amount of genomic and transcriptomic data currently available, we retrieved 51 SAA-like proteins in several protostome taxa comprising 21 marine bivalve species and basal metazoans. In addition to vertebrate-like SAAs, we identified a second protein type with peculiar features. In the bivalves Crassostrea gigas and Mytilus galloprovincialis, both digital expression analysis and qPCR data indicated an induction of the classical SAA after bacterial challenge.
Collapse
Affiliation(s)
- U Rosani
- Dept. of Biology, University of Padua, via U. Bassi 58/b, 35121 Padua, Italy
| | - S Domeneghetti
- Dept. of Biology, University of Padua, via U. Bassi 58/b, 35121 Padua, Italy
| | - M Gerdol
- Dept. of Life Sciences, University of Trieste, via L. Giorgeri 5, 34127 Trieste, Italy
| | - M Franzoi
- Dept. of Biology, University of Padua, via U. Bassi 58/b, 35121 Padua, Italy
| | - A Pallavicini
- Dept. of Life Sciences, University of Trieste, via L. Giorgeri 5, 34127 Trieste, Italy
| | - P Venier
- Dept. of Biology, University of Padua, via U. Bassi 58/b, 35121 Padua, Italy.
| |
Collapse
|
26
|
Trenchevska O, Nelson RW, Nedelkov D. Mass Spectrometric Immunoassays in Characterization of Clinically Significant Proteoforms. Proteomes 2016; 4:proteomes4010013. [PMID: 28248223 PMCID: PMC5217360 DOI: 10.3390/proteomes4010013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/10/2016] [Accepted: 03/14/2016] [Indexed: 02/07/2023] Open
Abstract
Proteins can exist as multiple proteoforms in vivo, as a result of alternative splicing and single-nucleotide polymorphisms (SNPs), as well as posttranslational processing. To address their clinical significance in a context of diagnostic information, proteoforms require a more in-depth analysis. Mass spectrometric immunoassays (MSIA) have been devised for studying structural diversity in human proteins. MSIA enables protein profiling in a simple and high-throughput manner, by combining the selectivity of targeted immunoassays, with the specificity of mass spectrometric detection. MSIA has been used for qualitative and quantitative analysis of single and multiple proteoforms, distinguishing between normal fluctuations and changes related to clinical conditions. This mini review offers an overview of the development and application of mass spectrometric immunoassays for clinical and population proteomics studies. Provided are examples of some recent developments, and also discussed are the trends and challenges in mass spectrometry-based immunoassays for the next-phase of clinical applications.
Collapse
Affiliation(s)
- Olgica Trenchevska
- The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA.
| | - Randall W Nelson
- The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA.
| | - Dobrin Nedelkov
- The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA.
| |
Collapse
|
27
|
Shahzad K, Akbar H, Vailati-Riboni M, Basiricò L, Morera P, Rodriguez-Zas S, Nardone A, Bernabucci U, Loor J. The effect of calving in the summer on the hepatic transcriptome of Holstein cows during the peripartal period. J Dairy Sci 2015; 98:5401-13. [DOI: 10.3168/jds.2015-9409] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 04/25/2015] [Indexed: 02/06/2023]
|
28
|
Jayasinghe JDHE, Elvitigala DAS, Whang I, Nam BH, Lee J. Molecular characterization of two immunity-related acute-phase proteins: Haptoglobin and serum amyloid A from black rockfish (Sebastes schlegeli). FISH & SHELLFISH IMMUNOLOGY 2015; 45:680-688. [PMID: 25989623 DOI: 10.1016/j.fsi.2015.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/04/2015] [Accepted: 05/10/2015] [Indexed: 06/04/2023]
Abstract
Haptoglobin (Hp) and serum amyloid A (SAA) are two vital proteins involved in inflammatory reactions and are classified as acute-phase proteins. They are released from hepatocytes under inflammatory conditions to protect healthy cells from being damaged by pathogens or from self-destructive mechanisms. In this study, a previously constructed black rockfish (Sebastes schlegeli) cDNA library was used to identify the full-length cDNA sequences of Hp and SAA homologs (RfHp and RfSAA, respectively) and characterize them at the molecular level. As expected, in silico analysis of these homologs showed the typical domain architectures of their known counterparts. Open reading frames of RfHp and RfSAA consisted of 942-bp and 313-bp DNA sequences, respectively. The derived polypeptide sequence of RfHp was composed of 313 amino acids (aa) with a predicted molecular weight of 34 kD, whereas RfSAA had a 121-amino acid sequence with a molecular weight of 13 kD. Phylogenetic analysis as well as pairwise sequence alignment results showed that RfHp was more closely related to Oreochromis mossambicus from an evolutionary perspective while RfSAA was closely related to the Epinephelus coioides ortholog. Although both genes were expressed ubiquitously in the tissues analyzed, they were particularly expressed in liver tissue, suggesting their origin in hepatocytes. Quantitative real-time PCR analysis indicated that both RfHp and RfSAA were significantly up-regulated by both bacterial and viral stimulation in liver tissue, affirming their putative importance in the acute phase of first-line host immune defenses.
Collapse
Affiliation(s)
- J D H E Jayasinghe
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Special Self-Governing Province 690-756, Republic of Korea
| | - Don Anushka Sandaruwan Elvitigala
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Special Self-Governing Province 690-756, Republic of Korea
| | - Ilson Whang
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Special Self-Governing Province 690-756, Republic of Korea
| | - Bo-Hye Nam
- Biotechnology Research Division, National Fisheries Research and Development Institute, 408-1 Sirang-ri, Gijang-up, Gijang-gun, Busan 619-705, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Special Self-Governing Province 690-756, Republic of Korea.
| |
Collapse
|
29
|
Jayaraman S, Haupt C, Gursky O. Thermal transitions in serum amyloid A in solution and on the lipid: implications for structure and stability of acute-phase HDL. J Lipid Res 2015; 56:1531-42. [PMID: 26022803 DOI: 10.1194/jlr.m059162] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Indexed: 12/20/2022] Open
Abstract
Serum amyloid A (SAA) is an acute-phase protein that circulates mainly on plasma HDL. SAA interactions with its functional ligands and its pathogenic deposition in reactive amyloidosis depend, in part, on the structural disorder of this protein and its propensity to oligomerize. In vivo, SAA can displace a substantial fraction of the major HDL protein, apoA-I, and thereby influence the structural remodeling and functions of acute-phase HDL in ways that are incompletely understood. We use murine SAA1.1 to report the first structural stability study of human plasma HDL that has been enriched with SAA. Calorimetric and spectroscopic analyses of these and other SAA-lipid systems reveal two surprising findings. First, progressive displacement of the exchangeable fraction of apoA-I by SAA has little effect on the structural stability of HDL and its fusion and release of core lipids. Consequently, the major determinant for HDL stability is the nonexchangeable apoA-I. A structural model explaining this observation is proposed, which is consistent with functional studies in acute-phase HDL. Second, we report an α-helix folding/unfolding transition in SAA in the presence of lipid at near-physiological temperatures. This new transition may have potentially important implications for normal functions of SAA and its pathogenic misfolding.
Collapse
Affiliation(s)
- Shobini Jayaraman
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston MA 02118
| | - Christian Haupt
- Institute for Pharmaceutical Biotechnology, University of Ulm, 89081, Ulm, Germany
| | - Olga Gursky
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston MA 02118
| |
Collapse
|
30
|
de Seny D, Cobraiville G, Charlier E, Neuville S, Lutteri L, Le Goff C, Malaise D, Malaise O, Chapelle JP, Relic B, Malaise MG. Apolipoprotein-A1 as a damage-associated molecular patterns protein in osteoarthritis: ex vivo and in vitro pro-inflammatory properties. PLoS One 2015; 10:e0122904. [PMID: 25849372 PMCID: PMC4388661 DOI: 10.1371/journal.pone.0122904] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/24/2015] [Indexed: 12/16/2022] Open
Abstract
Osteoarthritis (OA) is associated with a local inflammatory process. Dyslipidemia is known to be an underlying cause for the development of OA. Therefore, lipid and inflammatory levels were quantified ex vivo in blood and synovial fluid of OA patients (n=29) and compared to those of rheumatoid arthritis (RA) patients (n=27) or healthy volunteers (HV) (n=35). The role of apolipoprotein A-I (ApoA1) was investigated in vitro on inflammatory parameters using human joint cells isolated from cartilage and synovial membrane obtained from OA patients after joint replacement. Cells were stimulated with ApoA1 in the presence or not of serum amyloid A (SAA) protein and/or lipoproteins (LDL and HDL) at physiological concentration observed in OA synovial fluid. In our ex vivo study, ApoA1, LDL-C and total cholesterol levels were strongly correlated to each other inside the OA joint cavity whereas same levels were not or weakly correlated to their corresponding serum levels. In OA synovial fluid, ApoA1 was not as strongly correlated to HDL as observed in OA serum or in RA synovial fluid, suggesting a dissociative level between ApoA1 and HDL in OA synovial fluid. In vitro, ApoA1 induced IL-6, MMP-1 and MMP-3 expression by primary chondrocytes and fibroblast-like synoviocytes through TLR4 receptor. HDL and LDL attenuated joint inflammatory response induced by ApoA1 and SAA in a ratio dependent manner. In conclusion, a dysregulated lipidic profile in the synovial fluid of OA patients was observed and was correlated with inflammatory parameters in the OA joint cavity. Pro-inflammatory properties of ApoA1 were confirmed in vitro.
Collapse
Affiliation(s)
- Dominique de Seny
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Gaël Cobraiville
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Edith Charlier
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Sophie Neuville
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Laurence Lutteri
- Laboratory of Clinical Chemistry, CHU Hospital of Liege, 4000 Liège, Belgium
| | - Caroline Le Goff
- Laboratory of Clinical Chemistry, CHU Hospital of Liege, 4000 Liège, Belgium
| | - Denis Malaise
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Olivier Malaise
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Jean-Paul Chapelle
- Laboratory of Clinical Chemistry, CHU Hospital of Liege, 4000 Liège, Belgium
| | - Biserka Relic
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| | - Michel G. Malaise
- Laboratory of Rheumatology, GIGA Research, University of Liege and CHU Hospital of Liege, 4000 Liège, Belgium
| |
Collapse
|
31
|
Yassine HN, Trenchevska O, He H, Borges CR, Nedelkov D, Mack W, Kono N, Koska J, Reaven PD, Nelson RW. Serum amyloid a truncations in type 2 diabetes mellitus. PLoS One 2015; 10:e0115320. [PMID: 25607823 PMCID: PMC4301920 DOI: 10.1371/journal.pone.0115320] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 11/21/2014] [Indexed: 12/16/2022] Open
Abstract
Serum Amyloid A (SAA) is an acute phase protein complex consisting of several abundant isoforms. The N- terminus of SAA is critical to its function in amyloid formation. SAA is frequently truncated, either missing an arginine or an arginine-serine dipeptide, resulting in isoforms that may influence the capacity to form amyloid. However, the relative abundance of truncated SAA in diabetes and chronic kidney disease is not known.
Collapse
Affiliation(s)
- Hussein N Yassine
- University of Southern California, Los Angeles, CA, United States of America
| | | | - Huijuan He
- University of Southern California, Los Angeles, CA, United States of America
| | - Chad R Borges
- Arizona State University, Tempe, AZ, United States of America
| | - Dobrin Nedelkov
- Arizona State University, Tempe, AZ, United States of America
| | - Wendy Mack
- University of Southern California, Los Angeles, CA, United States of America
| | - Naoko Kono
- University of Southern California, Los Angeles, CA, United States of America
| | - Juraj Koska
- Phoenix VA Health Care System, Phoenix, AZ, United States of America
| | - Peter D Reaven
- Phoenix VA Health Care System, Phoenix, AZ, United States of America
| | | |
Collapse
|
32
|
Takase H, Furuchi H, Tanaka M, Yamada T, Matoba K, Iwasaki K, Kawakami T, Mukai T. Characterization of reconstituted high-density lipoprotein particles formed by lipid interactions with human serum amyloid A. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1842:1467-74. [PMID: 25063355 DOI: 10.1016/j.bbalip.2014.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 07/12/2014] [Accepted: 07/15/2014] [Indexed: 12/21/2022]
Abstract
The acute-phase human protein serum amyloid A (SAA) is enriched in high-density lipoprotein (HDL) in patients with inflammatory diseases. Compared with normal HDL containing apolipoprotein A-I, which is the principal protein component, characteristics of acute-phase HDL containing SAA remain largely undefined. In the present study, we examined the physicochemical properties of reconstituted HDL (rHDL) particles formed by lipid interactions with SAA. Fluorescence and circular dichroism measurements revealed that although SAA was unstructured at physiological temperature, α-helix formation was induced upon binding to phospholipid vesicles. SAA also formed rHDL particles by solubilizing phospholipid vesicles through mechanisms that are common to other exchangeable apolipoproteins. Dynamic light scattering and nondenaturing gradient gel electrophoresis analyses of rHDL after gel filtration revealed particle sizes of approximately 10nm, and a discoidal shape was verified by transmission electron microscopy. Thermal denaturation experiments indicated that SAA molecules in rHDL retained α-helical conformations at 37°C, but were almost completely denatured around 60°C. Furthermore, trypsin digestion experiments showed that lipid binding rendered SAA molecules resistant to protein degradation. In humans, three major SAA1 isoforms (SAA1.1, 1.3, and 1.5) are known. Although these isoforms have different amino acids at residues 52 and 57, no major differences in physicochemical properties between rHDL particles resulting from lipid interactions with SAA isoforms have been found. The present data provide useful insights into the effects of SAA enrichment on the physicochemical properties of HDL.
Collapse
Affiliation(s)
- Hiroka Takase
- Department of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe 658-8558, Japan
| | - Hiroki Furuchi
- Department of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe 658-8558, Japan
| | - Masafumi Tanaka
- Department of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe 658-8558, Japan.
| | - Toshiyuki Yamada
- Department of Clinical and Laboratory Medicine, Jichi Medical University, Tochigi 329-0498, Japan
| | - Kyoko Matoba
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita 565-0871, Japan
| | - Kenji Iwasaki
- Laboratory of Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita 565-0871, Japan
| | - Toru Kawakami
- Laboratory of Protein Organic Chemistry, Institute for Protein Research, Osaka University, Suita 565-0871, Japan
| | - Takahiro Mukai
- Department of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe 658-8558, Japan
| |
Collapse
|
33
|
Rossmann C, Hammer A, Koyani CN, Kovacevic A, Siwetz M, Desoye G, Poehlmann TG, Markert UR, Huppertz B, Sattler W, Malle E. Expression of serum amyloid A4 in human trophoblast-like choriocarcinoma cell lines and human first trimester/term trophoblast cells. Placenta 2014; 35:661-4. [PMID: 24951172 PMCID: PMC4119475 DOI: 10.1016/j.placenta.2014.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/27/2014] [Accepted: 05/29/2014] [Indexed: 11/16/2022]
Abstract
Trophoblast invasion into uterine tissues represents a hallmark of first trimester placental development. As expression of serum amyloid A4 (SAA4) occurs in tumorigenic and invasive tissues we here investigated whether SAA4 is present in trophoblast-like human AC1-M59/Jeg-3 cells and trophoblast preparations of human first trimester and term placenta. SAA4 mRNA was expressed in non-stimulated and cytokine-treated AC1-M59/Jeg-3 cells. In purified trophoblast cells SAA4 mRNA expression was upregulated at weeks 10 and 12 of pregnancy. Western-blot and immunohistochemical staining of first trimester placental tissue revealed pronounced SAA4 expression in invasive trophoblast cells indicating a potential role of SAA4 during invasion. SAA4 mRNA is expressed in Jeg-3 and AC1-M59 cells. SAA4 mRNA is expressed in first trimester/term trophoblast cells. SAA4 mRNA is upregulated at pregnancy week 10 and 12. SAA4 protein is present in interstitial, intramural and intraluminal trophoblast cells.
Collapse
Affiliation(s)
- C Rossmann
- Medical University of Graz, Institute of Molecular Biology and Biochemistry, Graz A-8010, Austria
| | - A Hammer
- Medical University of Graz, Institute of Cell Biology, Histology and Embryology, Graz, Austria
| | - C N Koyani
- Medical University of Graz, Institute of Molecular Biology and Biochemistry, Graz A-8010, Austria
| | - A Kovacevic
- Medical University of Graz, Institute of Molecular Biology and Biochemistry, Graz A-8010, Austria
| | - M Siwetz
- Medical University of Graz, Institute of Cell Biology, Histology and Embryology, Graz, Austria
| | - G Desoye
- Medical University of Graz, Department of Obstetrics and Gynecology, Graz, Austria
| | - T G Poehlmann
- Placenta-Laboratory, Department of Obstetrics, University Hospital Jena, Jena, Germany
| | - U R Markert
- Placenta-Laboratory, Department of Obstetrics, University Hospital Jena, Jena, Germany
| | - B Huppertz
- Medical University of Graz, Institute of Cell Biology, Histology and Embryology, Graz, Austria
| | - W Sattler
- Medical University of Graz, Institute of Molecular Biology and Biochemistry, Graz A-8010, Austria
| | - E Malle
- Medical University of Graz, Institute of Molecular Biology and Biochemistry, Graz A-8010, Austria.
| |
Collapse
|
34
|
Reisinger KW, Kramer BW, Van der Zee DC, Brouwers HAA, Buurman WA, van Heurn E, Derikx JPM. Non-invasive serum amyloid A (SAA) measurement and plasma platelets for accurate prediction of surgical intervention in severe necrotizing enterocolitis (NEC). PLoS One 2014; 9:e90834. [PMID: 24603723 PMCID: PMC3946234 DOI: 10.1371/journal.pone.0090834] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 02/05/2014] [Indexed: 12/02/2022] Open
Abstract
Objective To evaluate the value of biomarkers to detect severe NEC. Summary Background Data The time point of surgery in necrotizing enterocolitis (NEC) is critical. Therefore, there is a need for markers that detect severe NEC, because clinical signs of severe NEC often develop late. This study evaluated the value of biomarkers reflecting intestinal cell damage and inflammation to detect severe NEC. Methods 29 neonates with NEC were included. Two definitions of moderate versus severe NEC were analyzed: medical NEC (n = 12) versus surgical or fatal NEC (n = 17); and Bell stage II NEC (n = 13) versus stage III NEC (n = 16). Urinary intestinal fatty acid binding protein (I-FABP), serum amyloid A (SAA), C3a and C5a, and fecal calprotectin were measured. C-reactive protein (CRP), white blood cell count (WBC) and platelet count data were measured in blood. Results In both definitions of moderate versus severe NEC, urinary SAA levels were significantly higher in severe NEC. A cut-off value of 34.4 ng/ml was found in surgical NEC versus medical NEC (sensitivity, 83%; specificity, 83%; LR+, 4.88 (95% CI, 1.37–17.0); LR−, 0.20 (95% CI, 0.07–0.60)) at diagnosis of NEC and at one day prior to surgery in neonates who were operated later on. Combination of urinary SAA and platelet count increased the accuracy, with a sensitivity, 94%; specificity, 83%; LR+, 5.53 (95% CI, 1.57–20.0); and LR−, 0.07 (95% CI, 0.01–0.48). Conclusion Urinary SAA is an accurate marker in differentiating severe NEC from moderate NEC; particularly if combined with serum platelet count.
Collapse
Affiliation(s)
- Kostan W. Reisinger
- Department of Surgery, Maastricht University Medical Centre, and Nutrition and Toxicology Research Institute (NUTRIM), Maastricht, the Netherlands
- * E-mail:
| | - Boris W. Kramer
- Department of Pediatrics, Maastricht University Medical Centre, and School for Oncology and Developmental Biology (GROW), School of Mental Health and Neurosciences, Maastricht, the Netherlands
| | - David C. Van der Zee
- Department of Surgery, Wilhelmina Children’s Hospital, University Medical Centre, Utrecht, the Netherlands
| | - Hens A. A. Brouwers
- Department of Neonatology, Wilhelmina Children’s Hospital, University Medical Centre, Utrecht, the Netherlands
| | - Wim A. Buurman
- Formerly Department of Surgery, currently Maastricht University Medical Centre, NUTRIM Institute, Maastricht, the Netherlands
| | - Ernest van Heurn
- Department of Surgery, Maastricht University Medical Centre, and Nutrition and Toxicology Research Institute (NUTRIM), Maastricht, the Netherlands
| | - Joep P. M. Derikx
- Department of Surgery, Maastricht University Medical Centre, and Nutrition and Toxicology Research Institute (NUTRIM), Maastricht, the Netherlands
| |
Collapse
|
35
|
Spodzieja M, Rafalik M, Szymańska A, Kołodziejczyk AS, Czaplewska P. Interaction of serum amyloid A with human cystatin C--assessment of amino acid residues crucial for hCC-SAA formation (part II). J Mol Recognit 2014; 26:415-25. [PMID: 23836469 DOI: 10.1002/jmr.2283] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 04/29/2013] [Accepted: 04/30/2013] [Indexed: 11/07/2022]
Abstract
Secondary amyloid A (AA) amyloidosis is an important complication of some chronic inflammatory diseases, primarily rheumatoid arthritis (RA). It is a serious, potentially life-threatening disorder caused by the deposition of AA fibrils, which are derived from the circulatory, acute-phase-reactant, serum amyloid A protein (SAA). Recently, a specific interaction between SAA and the ubiquitous inhibitor of cysteine proteases--human cystatin C (hCC)--has been proved. Using a combination of selective proteolytic excision and high-resolution mass spectrometry, the binding sites in the SAA and hCC sequences were assessed as SAA(86-104) and hCC(96-102), respectively. Here, we report further details concerning the hCC-SAA interaction. With the use of affinity tests and florescent ELISA-like assays, the amino acid residues crucial for the protein interaction were determined. It was shown that all amino acid residues in the SAA sequence, essential for the formation of the protein complex, are basic ones, which suggests an electrostatic interaction character. The idea is corroborated by the fact that the most important residues in the hCC sequence are Ser-98 and Tyr-102; these residues are able to form hydrogen bonds via their hydroxyl groups. The molecular details of hCC-SAA complex formation might be helpful for the design of new compounds modulating the biological role of both proteins.
Collapse
Affiliation(s)
- Marta Spodzieja
- University of Gdansk, Department of Medicinal Chemistry, Gdansk, Poland
| | | | | | | | | |
Collapse
|
36
|
Hafiane A, Jabor B, Ruel I, Ling J, Genest J. High-density lipoprotein mediated cellular cholesterol efflux in acute coronary syndromes. Am J Cardiol 2014; 113:249-55. [PMID: 24210679 DOI: 10.1016/j.amjcard.2013.09.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 09/17/2013] [Accepted: 09/17/2013] [Indexed: 02/07/2023]
Abstract
Systemic inflammation at the development of an acute coronary syndrome (ACS) might alter the high-density lipoprotein (HDL) components and function. One of the major functions of HDL particles is their ability to remove cellular cholesterol from macrophages. The aim of the present study was to characterize the HDL efflux capacity in patients with ACS. We analyzed the cholesterol efflux in those ACS (within 72 hours of symptoms [ACS1]) and, again, 3 months later (ACS2). As controls, we used normal subjects without coronary artery disease (CAD) and patients with chronic, stable CAD. The 4 groups were matched for age and HDL cholesterol levels. We used a cell-based efflux system in (3)[H]-cholesterol-labeled J774 macrophages to measure cholesterol efflux from apolipoprotein B-depleted plasma. The present study included 20 patients with ACS. Their mean age was 58 ± 9 years, and the mean HDL cholesterol level was 1.06 ± 0.22 mmol/L (41 ± 9 mg/dl). The ACS1 group showed a marked increase in high-sensitivity C-reactive protein and serum amyloid A, reflecting systemic inflammation. The HDL cholesterol efflux capacity was reduced in ACS1 subjects and remained reduced 12 weeks later and in those with stable CAD. These results suggest that the acute presence of serum amyloid A does not account for the impairment of HDL-mediated cholesterol efflux capacity in the ACS1 group. Little correlation was found between HDL cholesterol and HDL efflux capacity (r = 0.233; p = 0.049), suggesting that HDL cholesterol is a poor marker of HDL function in inflammatory states and CAD. In conclusion, our data support the concept that atherogenic HDL dysfunction and impaired efflux occur in ACS, independent of changes in plasma HDL cholesterol and apolipoprotein A-I levels.
Collapse
Affiliation(s)
- Anouar Hafiane
- Cardiovascular Research Laboratories, Division of Cardiology, McGill University Faculty of Medicine, Royal Victoria Hospital, Montreal, Quebec, Canada
| | - Bashar Jabor
- Cardiovascular Research Laboratories, Division of Cardiology, McGill University Faculty of Medicine, Royal Victoria Hospital, Montreal, Quebec, Canada
| | - Isabelle Ruel
- Cardiovascular Research Laboratories, Division of Cardiology, McGill University Faculty of Medicine, Royal Victoria Hospital, Montreal, Quebec, Canada
| | - Jennifer Ling
- Cardiovascular Research Laboratories, Division of Cardiology, McGill University Faculty of Medicine, Royal Victoria Hospital, Montreal, Quebec, Canada
| | - Jacques Genest
- Cardiovascular Research Laboratories, Division of Cardiology, McGill University Faculty of Medicine, Royal Victoria Hospital, Montreal, Quebec, Canada.
| |
Collapse
|
37
|
Depletion of spleen macrophages delays AA amyloid development: a study performed in the rapid mouse model of AA amyloidosis. PLoS One 2013; 8:e79104. [PMID: 24236094 PMCID: PMC3827313 DOI: 10.1371/journal.pone.0079104] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 09/18/2013] [Indexed: 11/19/2022] Open
Abstract
AA amyloidosis is a systemic disease that develops secondary to chronic inflammatory diseases Macrophages are often found in the vicinity of amyloid deposits and considered to play a role in both formation and degradation of amyloid fibrils. In spleen reside at least three types of macrophages, red pulp macrophages (RPM), marginal zone macrophages (MZM), metallophilic marginal zone macrophages (MMZM). MMZM and MZM are located in the marginal zone and express a unique collection of scavenger receptors that are involved in the uptake of blood-born particles. The murine AA amyloid model that resembles the human form of the disease has been used to study amyloid effects on different macrophage populations. Amyloid was induced by intravenous injection of amyloid enhancing factor and subcutaneous injections of silver nitrate and macrophages were identified with specific antibodies. We show that MZMs are highly sensitive to amyloid and decrease in number progressively with increasing amyloid load. Total area of MMZMs is unaffected by amyloid but cells are activated and migrate into the white pulp. In a group of mice spleen macrophages were depleted by an intravenous injection of clodronate filled liposomes. Subsequent injections of AEF and silver nitrate showed a sustained amyloid development. RPMs that constitute the majority of macrophages in spleen, appear insensitive to amyloid and do not participate in amyloid formation.
Collapse
|
38
|
Samikkannu T, Rao KVK, Arias AY, Kalaichezian A, Sagar V, Yoo C, Nair MPN. HIV infection and drugs of abuse: role of acute phase proteins. J Neuroinflammation 2013; 10:113. [PMID: 24044608 PMCID: PMC3848479 DOI: 10.1186/1742-2094-10-113] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 09/04/2013] [Indexed: 01/12/2023] Open
Abstract
Background HIV infection and drugs of abuse such as methamphetamine (METH), cocaine, and alcohol use have been identified as risk factors for triggering inflammation. Acute phase proteins such as C-reactive protein (CRP) and serum amyloid A (SAA) are the biomarkers of inflammation. Hence, the interactive effect of drugs of abuse with acute phase proteins in HIV-positive subjects was investigated. Methods Plasma samples were utilized from 75 subjects with METH use, cocaine use, alcohol use, and HIV-positive alone and HIV-positive METH, cocaine, and alcohol users, and age-matched control subjects. The plasma CRP and SAA levels were measured by ELISA and western blot respectively and the CD4 counts were also measured. Results Observed results indicated that the CRP and SAA levels in HIV-positive subjects who are METH, cocaine and alcohol users were significantly higher when compared with either drugs of abuse or HIV-positive alone. The CD4 counts were also dramatically reduced in HIV-positive with drugs of abuse subjects compared with only HIV-positive subjects. Conclusions These results suggest that, in HIV-positive subjects, drugs of abuse increase the levels of CRP and SAA, which may impact on the HIV infection and disease progression.
Collapse
Affiliation(s)
- Thangavel Samikkannu
- Department of Immunology, Institute of NeuroImmune Pharmacology, College of Medicine, Florida International University, Miami, FL 33199, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Characterization of the oligomerization and aggregation of human Serum Amyloid A. PLoS One 2013; 8:e64974. [PMID: 23750222 PMCID: PMC3672174 DOI: 10.1371/journal.pone.0064974] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 04/19/2013] [Indexed: 01/26/2023] Open
Abstract
The fibrillation of Serum Amyloid A (SAA) – a major acute phase protein – is believed to play a role in the disease Amyloid A (AA) Amyloidosis. To better understand the amyloid formation pathway of SAA, we characterized the oligomerization, misfolding, and aggregation of a disease-associated isoform of human SAA – human SAA1.1 (hSAA1.1) – using techniques ranging from circular dichroism spectroscopy to atomic force microscopy, fluorescence spectroscopy, immunoblot studies, solubility measurements, and seeding experiments. We found that hSAA1.1 formed alpha helix-rich, marginally stable oligomers in vitro on refolding and cross-beta-rich aggregates following incubation at 37°C. Strikingly, while hSAA1.1 was not highly amyloidogenic in vitro, the addition of a single N-terminal methionine residue significantly enhanced the fibrillation propensity of hSAA1.1 and modulated its fibrillation pathway. A deeper understanding of the oligomerization and fibrillation pathway of hSAA1.1 may help elucidate its pathological role.
Collapse
|
40
|
Lee HY, Kim SD, Baek SH, Choi JH, Cho KH, Zabel BA, Bae YS. Serum amyloid A stimulates macrophage foam cell formation via lectin-like oxidized low-density lipoprotein receptor 1 upregulation. Biochem Biophys Res Commun 2013; 433:18-23. [PMID: 23454129 DOI: 10.1016/j.bbrc.2013.02.077] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 02/14/2013] [Indexed: 12/30/2022]
Abstract
Elevated levels of serum amyloid A (SAA) is a risk factor for cardiovascular diseases, however, the role of SAA in the pathophysiology of atherosclerosis remains unclear. Here we show that SAA induced macrophage foam cell formation. SAA-stimulated foam cell formation was mediated by c-jun N-terminal kinase (JNK) signaling. Moreover, both SAA and SAA-conjugated high density lipoprotein stimulated the expression of the important scavenger receptor lectin-like oxidized low-density lipoprotein receptor 1 (LOX1) via nuclear factor-κB (NF-κB). A LOX1 antagonist carrageenan significantly blocked SAA-induced foam cell formation, indicating that SAA promotes foam cell formation via LOX1 expression. Our findings therefore suggest that SAA stimulates foam cell formation via LOX1 induction, and thus likely contributes to atherogenesis.
Collapse
Affiliation(s)
- Ha Young Lee
- Department of Biological Science, Sungkyunkwan University, Suwon 440-746, South Korea.
| | | | | | | | | | | | | |
Collapse
|
41
|
Spodzieja M, Szymańska A, Kołodziejczyk A, Prądzińska M, Maszota M, Stefanowicz P, Szewczuk Z, Grubb A, Czaplewska P. Interaction of serum amyloid A with human cystatin C--identification of binding sites. J Mol Recognit 2013; 25:513-24. [PMID: 22996594 DOI: 10.1002/jmr.2220] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Serum amyloid A (SAA) is a multifunctional acute-phase protein whose natural role seems to be participation in many physiologic and pathological processes. Prolonged increased SAA level in a number of chronic inflammatory and neoplastic diseases gives rise to reactive systemic amyloid A amyloidosis, where the N-terminal 76-amino acid residue-long segment of SAA is deposited as amyloid fibrils. Recently, a specific interaction between SAA and the ubiquitous inhibitor of cysteine proteases--human cystatin C (hCC)--has been described. Here, we report further evidence corroborating this interaction, and the identification of the SAA and hCC binding sites in the SAA-hCC complex, using a combination of selective proteolytic excision and high-resolution mass spectrometry. The shortest binding site in the SAA sequence was determined as SAA(86-104), whereas the binding site in hCC sequence was identified as hCC(96-102). Binding specificities of both interacting sequences were ascertained by affinity experiments (ELISA) and by registration of mass spectrum of SAA-hCC complex.
Collapse
Affiliation(s)
- Marta Spodzieja
- Department of Medicinal Chemistry, University of Gdansk, Sobieskiego 18, 80-952 Gdansk, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Leow KY, Goh WWB, Tan SZ, Lim J, Ng K, Oh VMS, Low AFH, Heng CK. Variant screening of the serum amyloid A1 gene and functional study of the p.Gly90Asp variant for its role in atherosclerosis. Atherosclerosis 2013; 227:112-7. [PMID: 23357645 DOI: 10.1016/j.atherosclerosis.2013.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Revised: 12/29/2012] [Accepted: 01/01/2013] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Serum amyloid A1 (SAA1) is a major acute-phase protein that is increasingly used as a reliable predictor of coronary artery disease (CAD). In this study we aim to screen the SAAI promoter and exons for genetic variants and to determine their association with CAD. In addition, we also carried out functional study on a variant of p.Gly90Asp encoded by the SAA1 gene. METHODS Variant screening of SAA1 was performed using high resolution melting (HRM) analysis. Genetic association of p.Gly90Asp with CAD was determined in 800 CAD patients and 773 Chinese control subjects. Functional study of p.Gly90Asp was carried out using THP-1-derived macrophages and HL-60 promyelocytic leukemia cells. RESULTS A total of 6 SNPs were identified, of which 2 were found to be novel (c.-913G > A and c.92-5T > G). The rare allele of p.Gly90Asp has a lower frequency of 0.013 in the CAD patients although this is not statistically significant. Functional studies of p.Gly90Asp revealed that the variant has decreased upregulation of key cytokines such as IL-8, MCP-1 and TNF-α as well as SERPINB2. CONCLUSIONS We found the variant p.Gly90Asp SAA1 protein eliciting significantly reduced inflammatory responses in macrophages through a reduction in the secretion of inflammatory cytokines. Despite strong functional effects, the minor allele frequency is too low in the population to attain statistical significance difference between cases and controls.
Collapse
Affiliation(s)
- Koon-Yeow Leow
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E, Kent Ridge Rd, Singapore 119228, Singapore.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Autoantibodies to apolipoprotein A-1 in cardiovascular diseases: current perspectives. Clin Dev Immunol 2012; 2012:868251. [PMID: 23227091 PMCID: PMC3511844 DOI: 10.1155/2012/868251] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 10/19/2012] [Indexed: 01/31/2023]
Abstract
Immune-mediated inflammation plays a major role in atherosclerosis and atherothrombosis, two essential features for cardiovascular disease (CVD) development, currently considered as the leading cause of death in the western world. There is accumulating evidence showing that humoral autoimmunity might play an important role in CVD and that some autoantibodies could represent emerging cardiovascular risk factors. Recent studies demonstrate that IgG autoantibodies against apolipoprotein A-1 (apoA-1) are raised in many diseases associated with a high cardiovascular risk, such as systemic lupus erythematosus, acute coronary syndrome, rheumatoid arthritis, severe carotid stenosis, and end-stage renal disease. In this work, we aimed at reviewing current data in the literature pointing to anti-apolipoprotein A-1 antibodies (anti-apoA-1 IgG) as a possible prognostic and diagnostic biomarker of cardiovascular risk and appraising their potential role as active mediators of atherogenesis.
Collapse
|
44
|
Reisinger KW, Van der Zee DC, Brouwers HAA, Kramer BW, van Heurn LWE, Buurman WA, Derikx JPM. Noninvasive measurement of fecal calprotectin and serum amyloid A combined with intestinal fatty acid-binding protein in necrotizing enterocolitis. J Pediatr Surg 2012; 47:1640-5. [PMID: 22974599 DOI: 10.1016/j.jpedsurg.2012.02.027] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 02/18/2012] [Accepted: 02/18/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND Diagnosis of necrotizing enterocolitis (NEC), prevalent in premature infants, remains challenging. Enterocyte damage in NEC can be assessed by intestinal fatty acid-binding protein (I-FABP), with a sensitivity of 93% and a specificity of 90%. Numerous markers of inflammation are known, such as serum amyloid A (SAA) and fecal calprotectin. PURPOSE The aim of the present study was to evaluate which combination of noninvasive measurement of inflammatory markers and I-FABP improves the diagnostic accuracy in neonates suspected for NEC. METHODS In 62 neonates with clinical suspicion of NEC (29 with final diagnosis of NEC), urinary I-FABP, urinary SAA, and fecal calprotectin levels were determined quantitatively. Diagnostic accuracy was calculated for the combinations I-FABP-SAA and I-FABP-fecal calprotectin, using a multivariable logistic regression model. RESULTS The combination of SAA and I-FABP did not increase the diagnostic accuracy of I-FABP. However, the combination of fecal calprotectin and I-FABP improved accuracy significantly. The combination of urinary I-FABP and fecal calprotectin measurement produced a sensitivity of 94%, a specificity of 79%, a positive likelihood ratio of 4.48, and a negative likelihood ratio of 0.08. CONCLUSION The combination of noninvasive measurement of I-FABP and fecal calprotectin seems promising for diagnosing NEC at an early time point. Prospective analysis is required to confirm this finding and to evaluate better treatment strategies based on noninvasive measurement of I-FABP and calprotectin.
Collapse
Affiliation(s)
- Kostan W Reisinger
- Department of Surgery, Maastricht University Medical Center, and Nutrition and Toxicology Research Institute, 6200 MD, Maastricht, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
45
|
Ding Y, Yang L, Zhang S, Wang Y, Du Y, Pu J, Peng G, Chen Y, Zhang H, Yu J, Hang H, Wu P, Yang F, Yang H, Steinbüchel A, Liu P. Identification of the major functional proteins of prokaryotic lipid droplets. J Lipid Res 2012; 53:399-411. [PMID: 22180631 PMCID: PMC3276463 DOI: 10.1194/jlr.m021899] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Revised: 12/15/2011] [Indexed: 11/20/2022] Open
Abstract
Storage of cellular triacylglycerols (TAGs) in lipid droplets (LDs) has been linked to the progression of many metabolic diseases in humans, and to the development of biofuels from plants and microorganisms. However, the biogenesis and dynamics of LDs are poorly understood. Compared with other organisms, bacteria seem to be a better model system for studying LD biology, because they are relatively simple and are highly efficient in converting biomass to TAG. We obtained highly purified LDs from Rhodococcus sp. RHA1, a bacterium that can produce TAG from many carbon sources, and then comprehensively characterized the LD proteome. Of the 228 LD-associated proteins identified, two major proteins, ro02104 and PspA, constituted about 15% of the total LD protein. The structure predicted for ro02104 resembles that of apolipoproteins, the structural proteins of plasma lipoproteins in mammals. Deletion of ro02104 resulted in the formation of supersized LDs, indicating that ro02104 plays a critical role in cellular LD dynamics. The putative α helix of the ro02104 LD-targeting domain (amino acids 83-146) is also similar to that of apolipoproteins. We report the identification of 228 proteins in the proteome of prokaryotic LDs, identify a putative structural protein of this organelle, and suggest that apolipoproteins may have an evolutionarily conserved role in the storage and trafficking of neutral lipids.
Collapse
Affiliation(s)
- Yunfeng Ding
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China; National Laboratory of Biomacromolecules, Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Li Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China; National Laboratory of Biomacromolecules, Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Shuyan Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China
| | - Yang Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China; National Laboratory of Biomacromolecules, Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Yalan Du
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China; National Laboratory of Biomacromolecules, Department of Histology and Embryology, University of South China, Hengyang, Hunan Province, China
| | - Jing Pu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China; National Laboratory of Biomacromolecules, Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Gong Peng
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China; National Laboratory of Biomacromolecules, Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Yong Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China
| | - Huina Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China
| | - Jinhai Yu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China; National Laboratory of Biomacromolecules, Graduate University of Chinese Academy of Sciences, Beijing, China
| | - Haiying Hang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China
| | - Peng Wu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China
| | - Fuquan Yang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China
| | - Hongyuan Yang
- National Laboratory of Biomacromolecules, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Alexander Steinbüchel
- National Laboratory of Biomacromolecules, Institut für Molekulare Mikrobiologie und Biotechnologie, Westfälische Wilhelms-Universität, D-48149 Münster, Germany, and King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Beijing, China.
| |
Collapse
|
46
|
Leow KY, Goh WWB, Heng CK. Effect of serum amyloid A1 treatment on global gene expression in THP-1-derived macrophages. Inflamm Res 2012; 61:391-8. [PMID: 22228103 DOI: 10.1007/s00011-011-0424-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 11/29/2011] [Accepted: 12/19/2011] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To investigate the effect of serum amyloid A1 (SAA1) on global gene expression in macrophages derived from THP-1 monocytes. MATERIALS AND METHODS Global genetic expression in THP-1-derived macrophages was determined using Illumina HT-12 microarray chips and the results were validated by real-time PCR. Cytokine levels in cellular supernatant were quantified by ELISA. RESULTS In total, 55 genes were upregulated with fold difference greater than two when THP-1-derived macrophages were incubated with SAA1 for 8 h. SAA1 is a strong cytokine inducer with significant upregulation of chemokines CCL1, CCL3, and CCL4 and this was confirmed by both real-time PCR and ELISA quantification. SAA1 also promotes the upregulation of genes involved in phagocytosis, anti-apoptosis, and tissue remodeling. CONCLUSIONS SAA1 appears to play an important role during the immune response and in chronic inflammatory diseases through the stimulation of genes involved in cytokine production, phagocytosis, and anti-apoptosis.
Collapse
Affiliation(s)
- Koon-Yeow Leow
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E, Kent Ridge Road, Singapore 119228, Singapore
| | | | | |
Collapse
|
47
|
Shores DR, Binion DG, Freeman BA, Baker PR. New insights into the role of fatty acids in the pathogenesis and resolution of inflammatory bowel disease. Inflamm Bowel Dis 2011; 17:2192-204. [PMID: 21910181 PMCID: PMC4100336 DOI: 10.1002/ibd.21560] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 10/05/2010] [Indexed: 12/12/2022]
Abstract
Dietary and endogenously modified lipids modulate inflammation by functioning as intra- and intercellular signaling molecules. Proinflammatory lipid mediators such as the eicosanoids compete against the signaling actions of newly discovered modified fatty acids that act to resolve inflammation. In inflammatory bowel disease, multiple aberrancies in lipid metabolism have been discovered, which shed further light on the pathogenesis of intestinal inflammation. Mechanisms by which lipids modulate inflammation, abnormalities of lipid metabolism in the setting of inflammatory bowel disease, and potential therapeutic application of lipid derivatives in this setting are discussed.
Collapse
Affiliation(s)
- Darla R. Shores
- Division of Pediatric Gastroenterology, Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania
| | - David G. Binion
- Division of Gastroenterology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Bruce A. Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Paul R.S. Baker
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
48
|
Cetinkaya M, Ozkan H, Köksal N, Akaci O, Ozgür T. Comparison of the efficacy of serum amyloid A, C-reactive protein, and procalcitonin in the diagnosis and follow-up of necrotizing enterocolitis in premature infants. J Pediatr Surg 2011; 46:1482-9. [PMID: 21843712 DOI: 10.1016/j.jpedsurg.2011.03.069] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2010] [Revised: 02/07/2011] [Accepted: 03/16/2011] [Indexed: 11/25/2022]
Abstract
PURPOSE The aim of this study was to compare the efficacy of serum amyloid A (SAA) with that of C-reactive protein (CRP), and procalcitonin (PCT) in diagnosis and follow-up of necrotizing enterocolitis (NEC) in preterm infants. METHODS A total of 152 infants were enrolled into this observational study. The infants were classified into 3 groups: group 1 (58 infants with NEC and sepsis), group 2 (54 infants with only sepsis), and group 3 (40 infants with neither sepsis nor NEC, or control group). The data including whole blood count, CRP, PCT, SAA, and cultures that were obtained at diagnosis (0 hour), at 24 and 48 hours, and at 7 and 10 days were evaluated. RESULTS A total of 58 infants had a diagnosis of NEC. Mean CRP (7.4 ± 5.2 mg/dL) and SAA (46.2 ± 41.3 mg/dL) values of infants in group 1 at 0 hour were significantly higher than those in groups 2 and 3. Although the area under the curve of CRP was higher at 0 hour in infants with NEC, there were no significant differences between groups with respect to the areas under the curve of SAA, CRP, and PCT at all measurement times. Levels of SAA decreased earlier than CRP and PCT in the follow-up of NEC (mean SAA levels were 45.8 ± 45.2, 21.9 ± 16.6, 10.1 ± 8.3, and 7.9 ± 5.1 mg/dL at evaluation times, respectively). Levels of CRP and SAA of infants with NEC stages II and III were significantly higher than those with only sepsis and/or NEC stage I. CONCLUSIONS Serum amyloid A, CRP, and PCT all are accurate and reliable markers in diagnosis of NEC, in addition to clinical and radiographic findings. Higher CRP and SAA levels might indicate advanced stage of NEC. Serial measurements of SAA, CRP, and PCT, either alone or in combination, can be used safely in the diagnosis and follow-up of NEC.
Collapse
Affiliation(s)
- Merih Cetinkaya
- Department of Neonatology, School of Medicine, Uludag University, Bursa, Turkey.
| | | | | | | | | |
Collapse
|
49
|
Chiba T, Chang MY, Wang S, Wight TN, McMillen TS, Oram JF, Vaisar T, Heinecke JW, De Beer FC, De Beer MC, Chait A. Serum amyloid A facilitates the binding of high-density lipoprotein from mice injected with lipopolysaccharide to vascular proteoglycans. Arterioscler Thromb Vasc Biol 2011; 31:1326-32. [PMID: 21474830 DOI: 10.1161/atvbaha.111.226159] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Levels of serum amyloid A (SAA), an acute-phase protein carried on high-density lipoprotein (HDL), increase in inflammatory states and are associated with increased risk of cardiovascular disease. HDL colocalizes with vascular proteoglycans in atherosclerotic lesions. However, its major apolipoprotein, apolipoprotein A-I, has no proteoglycan-binding domains. Therefore, we investigated whether SAA, which has proteoglycan-binding domains, plays a role in HDL retention by proteoglycans. METHODS AND RESULTS HDL from control mice and mice deficient in both SAA1.1 and SAA2.1 (SAA knockout mice) injected with bacterial lipopolysaccharide (LPS) was studied. SAA mRNA expression in the liver and plasma levels of SAA increased dramatically in C57BL/6 mice after LPS administration, although HDL cholesterol did not change. Fast protein liquid chromatography analysis showed most of the SAA to be in HDL. Mass spectrometric analysis indicated that HDL from LPS-injected control mice had high levels of SAA1.1/2.1 and reduced levels of apolipoprotein A-I. HDL from LPS-injected control mice demonstrated high-affinity binding to biglycan relative to normal mouse HDL. In contrast, HDL from LPS-injected SAA knockout mice showed very little binding to biglycan, consistent with SAA facilitating the binding of HDL to vascular proteoglycans. CONCLUSION SAA enrichment of HDL under inflammatory conditions plays an important role in the binding of HDL to vascular proteoglycans.
Collapse
Affiliation(s)
- Tsuyoshi Chiba
- Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tuomainen AM, Hyvärinen K, Ehlers PI, Mervaala E, Leinonen M, Saikku P, Kovanen PT, Jauhiainen M, Pussinen PJ. The effect of proatherogenic microbes on macrophage cholesterol homeostasis in apoE-deficient mice. Microb Pathog 2011; 51:217-24. [PMID: 21421042 DOI: 10.1016/j.micpath.2011.03.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 03/11/2011] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pathogens such as Aggregatibacter actinomycetemcomitans (Aa) and Chlamydia pneumoniae (Cpn) associate with an increased risk for cardiovascular diseases by inducing inflammation. We hypothesized that the pathogens affect the vascular wall by disturbing cholesterol homeostasis and endothelial function. METHODS Aa- and Cpn-infections were induced in apoE-deficient mice by intravenous and intranasal applications, respectively. Cholesterol efflux from mouse peritoneal macrophages to apo(lipoprotein)A-I was assessed. The efflux capacity of mouse sera as acceptors of cholesterol from RAW264.7-macrophages was determined. Additionally, endothelial function was studied by following the relaxation capacity of rat mesenteric arteries after incubation in the conditioned culture media of the peritoneal macrophages isolated from the mice. RESULTS Infection increased serum phospholipid transfer protein (PLTP) and lipopolysaccharide (LPS) activity, as well as serum amyloid A (SAA) and TNF-α concentrations. Peritoneal macrophages of mice with Aa-infection showed increased cholesterol uptake and reduced cholesterol efflux. Sera of Cpn and Cpn + Aa-infected mice had reduced cholesterol efflux capacity from RAW264.7-macrophages. Conditioned macrophage medium from mice with chronic C. pneumoniae infection induced endothelial dysfunction. Additionally, concentrations of serum adhesion molecules, intercellular adhesion molecule (ICAM) and vascular cell adhesion molecule (VCAM) in Cpn-groups and E-selectin in Cpn + Aa-group, were elevated. The serum markers of endothelial function correlated positively with SAA. CONCLUSIONS Aa- and Cpn-infections may generate proatherogenic changes in the vascular wall by affecting the macrophage cholesterol homeostasis and endothelial function.
Collapse
Affiliation(s)
- Anita M Tuomainen
- Institute of Dentistry, University of Helsinki, FI-00014 Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|