1
|
Dayer R, De Marco G, Vazquez O, Tabard-Fougère A, Cochard B, Gavira N, Di Laura Frattura G, Guanziroli Pralong N, Steiger C, Ceroni D. Laboratory diagnostics for primary spinal infections in pediatric and adult populations: a narrative review. NORTH AMERICAN SPINE SOCIETY JOURNAL 2023; 16:100270. [PMID: 37767011 PMCID: PMC10520565 DOI: 10.1016/j.xnsj.2023.100270] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/02/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023]
Abstract
Primary spinal infection (PSI) is a generic term covering a heterogeneous group of infections that can affect the vertebral body, intervertebral disks, the content of the medullary cavity, and adjacent paraspinal tissues. Patients' characteristics can vary significantly, notably according to their age, and some of these characteristics undoubtedly play a primordial role in the occurrence of a PSI and in the type of offending pathogen. Before approaching the subject of laboratory diagnostics, it is essential to define the characteristics of the patient and their infection, which can then guide the physician toward specific diagnostic approaches. This review critically examined the roles and usefulness of traditional and modern laboratory diagnostics in supporting clinicians' decision-making in cases of pediatric and adult primary spinal infection (PSI). It appears impossible to compare PSIs in children and adults, whether from an epidemiological, clinical, bacteriological, or biological perspective. The recipients are really too different, and the responsible germs are closely correlated to their age. Secondly, the interpretation of traditional laboratory blood tests appears to contribute little guidance for clinicians attempting to diagnose a PSI. Biopsy or needle aspiration for bacterial identification remains a controversial subject, as the success rates of these procedures for identifying causative organisms are relatively uncertain in pediatric populations.Using nucleic acid amplification assays (NAAAs) on biopsy samples has been demonstrated to be more sensitive than conventional cultures for diagnosing PSI. Recent advances in next-generation sequencing (NGS) are particularly interesting for establishing a microbiological diagnosis of a PSI when standard cultures and NAAAs have failed to detect the culprit. We can even imagine that plasma metagenomic NGS using plasma (known as "liquid biopsy") is a diagnostic approach that can detect not only pathogens circulating in the bloodstream but also those causing focal infections, and thus eliminate the need for source sample collection using costly invasive surgical procedures.
Collapse
Affiliation(s)
- Romain Dayer
- Pediatric Orthopedics Unit, Pediatric Surgery Service, Geneva University Hospitals and University of Geneva Rue Willy-Donzé 6, Geneva 1205, Switzerland
| | - Giacomo De Marco
- Pediatric Orthopedics Unit, Pediatric Surgery Service, Geneva University Hospitals and University of Geneva Rue Willy-Donzé 6, Geneva 1205, Switzerland
| | - Oscar Vazquez
- Pediatric Orthopedics Unit, Pediatric Surgery Service, Geneva University Hospitals and University of Geneva Rue Willy-Donzé 6, Geneva 1205, Switzerland
| | - Anne Tabard-Fougère
- Pediatric Orthopedics Unit, Pediatric Surgery Service, Geneva University Hospitals and University of Geneva Rue Willy-Donzé 6, Geneva 1205, Switzerland
| | - Blaise Cochard
- Pediatric Orthopedics Unit, Pediatric Surgery Service, Geneva University Hospitals and University of Geneva Rue Willy-Donzé 6, Geneva 1205, Switzerland
| | - Nathaly Gavira
- Pediatric Orthopedics Unit, Pediatric Surgery Service, Geneva University Hospitals and University of Geneva Rue Willy-Donzé 6, Geneva 1205, Switzerland
| | - Giorgio Di Laura Frattura
- Pediatric Orthopedics Unit, Pediatric Surgery Service, Geneva University Hospitals and University of Geneva Rue Willy-Donzé 6, Geneva 1205, Switzerland
| | - Nastassia Guanziroli Pralong
- Pediatric Orthopedics Unit, Pediatric Surgery Service, Geneva University Hospitals and University of Geneva Rue Willy-Donzé 6, Geneva 1205, Switzerland
| | - Christina Steiger
- Pediatric Orthopedics Unit, Pediatric Surgery Service, Geneva University Hospitals and University of Geneva Rue Willy-Donzé 6, Geneva 1205, Switzerland
| | - Dimitri Ceroni
- Pediatric Orthopedics Unit, Pediatric Surgery Service, Geneva University Hospitals and University of Geneva Rue Willy-Donzé 6, Geneva 1205, Switzerland
| |
Collapse
|
2
|
Toobian D, Ghosh P, Katkar GD. Parsing the Role of PPARs in Macrophage Processes. Front Immunol 2021; 12:783780. [PMID: 35003101 PMCID: PMC8727354 DOI: 10.3389/fimmu.2021.783780] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Cells are richly equipped with nuclear receptors, which act as ligand-regulated transcription factors. Peroxisome proliferator activated receptors (PPARs), members of the nuclear receptor family, have been extensively studied for their roles in development, differentiation, and homeostatic processes. In the recent past, there has been substantial interest in understanding and defining the functions of PPARs and their agonists in regulating innate and adaptive immune responses as well as their pharmacologic potential in combating acute and chronic inflammatory disease. In this review, we focus on emerging evidence of the potential roles of the PPAR subtypes in macrophage biology. We also discuss the roles of dual and pan PPAR agonists as modulators of immune cell function, microbial infection, and inflammatory diseases.
Collapse
Affiliation(s)
- Daniel Toobian
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, United States
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, United States
- Rebecca and John Moore Comprehensive Cancer Center, University of California San Diego, San Diego, CA, United States
- Department of Medicine, University of California San Diego, San Diego, CA, United States
- Veterans Affairs Medical Center, La Jolla, CA, United States
| | - Gajanan D. Katkar
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
3
|
Monophosphoryl Lipid A Enhances Efficacy of a Francisella tularensis LVS-Catanionic Nanoparticle Subunit Vaccine against F. tularensis Schu S4 Challenge by Augmenting both Humoral and Cellular Immunity. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00574-16. [PMID: 28077440 DOI: 10.1128/cvi.00574-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/06/2017] [Indexed: 01/01/2023]
Abstract
Francisella tularensis, a bacterial biothreat agent, has no approved vaccine in the United States. Previously, we showed that incorporating lysates from partially attenuated F. tularensis LVS or fully virulent F. tularensis Schu S4 strains into catanionic surfactant vesicle (V) nanoparticles (LVS-V and Schu S4-V, respectively) protected fully against F. tularensis LVS intraperitoneal (i.p.) challenge in mice. However, we achieved only partial protection against F. tularensis Schu S4 intranasal (i.n.) challenge, even when employing heterologous prime-boost immunization strategies. We now extend these findings to show that both LVS-V and Schu S4-V immunization (i.p./i.p.) elicited similarly high titers of anti-F. tularensis IgG and that the titers could be further increased by adding monophosphoryl lipid A (MPL), a nontoxic Toll-like receptor 4 (TLR4) adjuvant that is included in several U.S. FDA-approved vaccines. LVS-V+MPL immune sera also detected more F. tularensis antigens than LVS-V immune sera and, after passive transfer to naive mice, significantly delayed the time to death against F. tularensis Schu S4 subcutaneous (s.c.) but not i.n. challenge. Active immunization with LVS-V+MPL (i.p./i.p.) also increased the frequency of gamma interferon (IFN-γ)-secreting activated helper T cells, IFN-γ production, and the ability of splenocytes to control intramacrophage F. tularensis LVS replication ex vivo Active LVS-V+MPL immunization via heterologous routes (i.p./i.n.) significantly elevated IgA and IgG levels in bronchoalveolar lavage fluid and significantly enhanced protection against i.n. F. tularensis Schu S4 challenge (to ∼60%). These data represent a significant step in the development of a subunit vaccine against the highly virulent type A strains.
Collapse
|
4
|
Development of a Multivalent Subunit Vaccine against Tularemia Using Tobacco Mosaic Virus (TMV) Based Delivery System. PLoS One 2015; 10:e0130858. [PMID: 26098553 PMCID: PMC4476615 DOI: 10.1371/journal.pone.0130858] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/26/2015] [Indexed: 11/21/2022] Open
Abstract
Francisella tularensis is a facultative intracellular pathogen, and is the causative agent of a fatal human disease known as tularemia. F. tularensis is classified as a Category A Biothreat agent by the CDC based on its use in bioweapon programs by several countries in the past and its potential to be used as an agent of bioterrorism. No licensed vaccine is currently available for prevention of tularemia. In this study, we used a novel approach for development of a multivalent subunit vaccine against tularemia by using an efficient tobacco mosaic virus (TMV) based delivery platform. The multivalent subunit vaccine was formulated to contain a combination of F. tularensis protective antigens: OmpA-like protein (OmpA), chaperone protein DnaK and lipoprotein Tul4 from the highly virulent F. tularensis SchuS4 strain. Two different vaccine formulations and immunization schedules were used. The immunized mice were challenged with lethal (10xLD100) doses of F. tularensis LVS on day 28 of the primary immunization and observed daily for morbidity and mortality. Results from this study demonstrate that TMV can be used as a carrier for effective delivery of multiple F. tularensis antigens. TMV-conjugate vaccine formulations are safe and multiple doses can be administered without causing any adverse reactions in immunized mice. Immunization with TMV-conjugated F. tularensis proteins induced a strong humoral immune response and protected mice against respiratory challenges with very high doses of F. tularensis LVS. This study provides a proof-of-concept that TMV can serve as a suitable platform for simultaneous delivery of multiple protective antigens of F. tularensis. Refinement of vaccine formulations coupled with TMV-targeting strategies developed in this study will provide a platform for development of an effective tularemia subunit vaccine as well as a vaccination approach that may broadly be applicable to many other bacterial pathogens.
Collapse
|
5
|
Oshida K, Vasani N, Thomas RS, Applegate D, Rosen M, Abbott B, Lau C, Guo G, Aleksunes LM, Klaassen C, Corton JC. Identification of modulators of the nuclear receptor peroxisome proliferator-activated receptor α (PPARα) in a mouse liver gene expression compendium. PLoS One 2015; 10:e0112655. [PMID: 25689681 PMCID: PMC4331523 DOI: 10.1371/journal.pone.0112655] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/09/2014] [Indexed: 12/22/2022] Open
Abstract
The nuclear receptor family member peroxisome proliferator-activated receptor α (PPARα) is activated by therapeutic hypolipidemic drugs and environmentally-relevant chemicals to regulate genes involved in lipid transport and catabolism. Chronic activation of PPARα in rodents increases liver cancer incidence, whereas suppression of PPARα activity leads to hepatocellular steatosis. Analytical approaches were developed to identify biosets (i.e., gene expression differences between two conditions) in a genomic database in which PPARα activity was altered. A gene expression signature of 131 PPARα-dependent genes was built using microarray profiles from the livers of wild-type and PPARα-null mice after exposure to three structurally diverse PPARα activators (WY-14,643, fenofibrate and perfluorohexane sulfonate). A fold-change rank-based test (Running Fisher’s test (p-value ≤ 10-4)) was used to evaluate the similarity between the PPARα signature and a test set of 48 and 31 biosets positive or negative, respectively for PPARα activation; the test resulted in a balanced accuracy of 98%. The signature was then used to identify factors that activate or suppress PPARα in an annotated mouse liver/primary hepatocyte gene expression compendium of ~1850 biosets. In addition to the expected activation of PPARα by fibrate drugs, di(2-ethylhexyl) phthalate, and perfluorinated compounds, PPARα was activated by benzofuran, galactosamine, and TCDD and suppressed by hepatotoxins acetaminophen, lipopolysaccharide, silicon dioxide nanoparticles, and trovafloxacin. Additional factors that activate (fasting, caloric restriction) or suppress (infections) PPARα were also identified. This study 1) developed methods useful for future screening of environmental chemicals, 2) identified chemicals that activate or suppress PPARα, and 3) identified factors including diets and infections that modulate PPARα activity and would be hypothesized to affect chemical-induced PPARα activity.
Collapse
Affiliation(s)
- Keiyu Oshida
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
| | - Naresh Vasani
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
| | - Russell S. Thomas
- Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina, United States of America
| | - Dawn Applegate
- RegeneMed, San Diego, California, United States of America
| | - Mitch Rosen
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
| | - Barbara Abbott
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
| | - Christopher Lau
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
| | - Grace Guo
- Rutgers University, Ernest Mario School of Pharmacy, Department of Pharmacology and Toxicology, Piscataway, New Jersey, United States of America
| | - Lauren M. Aleksunes
- Rutgers University, Ernest Mario School of Pharmacy, Department of Pharmacology and Toxicology, Piscataway, New Jersey, United States of America
| | - Curtis Klaassen
- University of Washington, Seattle, Washington, United States of America
| | - J. Christopher Corton
- National Health and Environmental Effects Research Lab, US-EPA, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
6
|
Mandard S, Patsouris D. Nuclear control of the inflammatory response in mammals by peroxisome proliferator-activated receptors. PPAR Res 2013; 2013:613864. [PMID: 23577023 PMCID: PMC3614066 DOI: 10.1155/2013/613864] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 01/14/2013] [Accepted: 01/29/2013] [Indexed: 12/30/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that play pivotal roles in the regulation of a very large number of biological processes including inflammation. Using specific examples, this paper focuses on the interplay between PPARs and innate immunity/inflammation and, when possible, compares it among species. We focus on recent discoveries establishing how inflammation and PPARs interact in the context of obesity-induced inflammation and type 2 diabetes, mostly in mouse and humans. We illustrate that PPAR γ ability to alleviate obesity-associated inflammation raises an interesting pharmacologic potential. In the light of recent findings, the protective role of PPAR α and PPAR β / δ against the hepatic inflammatory response is also addressed. While PPARs agonists are well-established agents that can treat numerous inflammatory issues in rodents and humans, surprisingly very little has been described in other species. We therefore also review the implication of PPARs in inflammatory bowel disease; acute-phase response; and central, cardiac, and endothelial inflammation and compare it along different species (mainly mouse, rat, human, and pig). In the light of the data available in the literature, there is no doubt that more studies concerning the impact of PPAR ligands in livestock should be undertaken because it may finally raise unconsidered health and sanitary benefits.
Collapse
Affiliation(s)
- Stéphane Mandard
- Centre de Recherche INSERM-UMR866 “Lipides, Nutrition, Cancer” Faculté de Médecine, Université de Bourgogne 7, Boulevard Jeanne d'Arc, 21079 Dijon Cedex, France
| | - David Patsouris
- Laboratoire CarMeN, UMR INSERM U1060/INRA 1235, Université Lyon 1, Faculté de Médecine Lyon Sud, 165 Chemin du Grand Revoyet, 69921 Oullins, France
- Department of Chemical Physiology, The Scripps Research Institute, MB-24, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
7
|
Pohanka M, Pavlis O, Ruttkay-Nedecky B, Sochor J, Sobotka J, Pikula J, Adam V, Kizek R. Tularemia progression accompanied with oxidative stress and antioxidant alteration in spleen and liver of BALB/c mice. J Microbiol 2012; 50:401-8. [PMID: 22752903 DOI: 10.1007/s12275-012-1621-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 01/17/2012] [Indexed: 10/28/2022]
Abstract
Francisella tularensis is the causative agent of tularemia. It is an intracellular pathogen with the ability to survive within phagosomes and induce pyroptotic cell death. In this study, we attempted to prove whether oxidative imbalance plays a significant role in tularemia pathogenesis. In our experimental model, we subcutaneously infected female BALB/c mice (dose 10(5) CFU of F. tularensis LVS). Liver, spleen, and blood were collected from mice at regular intervals from days 1-15 after infection. The bacterial burden was assessed by a cultivation test. The burden was unchanging from the 2(nd) to 6(th) day after infection. The bacterial burden corresponded to the plasmatic level of IFN-γ, IL-6, and liver malondialdehyde. After the phase of acute bacteraemia and the innate immunity reaction, the levels of reduced glutathione and total low molecular weight antioxidants decreased significantly and the activity of caspase-3 increased in the liver. The level of reduced glutathione decreased to 25% of the original level, and the total level of low molecular weight antioxidants was less than 50% of the initial amount. The demonstrated effects of tularemia-induced pathology had a more extensive impact on the liver than on the spleen.
Collapse
Affiliation(s)
- Miroslav Pohanka
- Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 500 01, Hradec Kralove, Czech Republic.
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Downer EJ, Clifford E, Amu S, Fallon PG, Moynagh PN. The synthetic cannabinoid R(+)WIN55,212-2 augments interferon-β expression via peroxisome proliferator-activated receptor-α. J Biol Chem 2012; 287:25440-53. [PMID: 22654113 DOI: 10.1074/jbc.m112.371757] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have demonstrated that R(+)WIN55,212-2, a synthetic cannabinoid that possesses cannabimimetic properties, acts as a novel regulator of Toll-like receptor 3 (TLR3) signaling to interferon (IFN) regulatory factor 3 (IRF3) activation and IFN-β expression, and this is critical for manifesting its protective effects in a murine multiple sclerosis model. Here we investigated the role of peroxisome proliferator-activated receptor-α (PPARα) in mediating the effects of R(+)WIN55,212-2 on this pathway. Data herein demonstrate that the TLR3 agonist poly(I:C) promotes IFN-β expression and R(+)WIN55,212-2 enhances TLR3-induced IFN-β expression in a stereoselective manner via PPARα. R(+)WIN55,212-2 promotes increased transactivation and expression of PPARα. Using the PPARα antagonist GW6471, we demonstrate that R(+)WIN55,212-2 acts via PPARα to activate JNK, activator protein-1, and positive regulatory domain IV to transcriptionally regulate the IFN-β promoter. Furthermore, GW6471 ameliorated the protective effects of R(+)WIN55,212-2 during the initial phase of experimental autoimmune encephalomyelitis. Overall, these findings define PPARα as an important mediator in manifesting the effects of R(+)WIN55,212-2 on the signaling cascade regulating IFN-β expression. The study adds to our molecular appreciation of potential therapeutic effects of R(+)WIN55,212-2 in multiple sclerosis.
Collapse
Affiliation(s)
- Eric J Downer
- Institute of Immunology, National University of Ireland Maynooth, County Kildare, Ireland
| | | | | | | | | |
Collapse
|
9
|
|
10
|
Guri AJ, Mohapatra SK, Horne WT, Hontecillas R, Bassaganya-Riera J. The role of T cell PPAR gamma in mice with experimental inflammatory bowel disease. BMC Gastroenterol 2010; 10:60. [PMID: 20537136 PMCID: PMC2891618 DOI: 10.1186/1471-230x-10-60] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Accepted: 06/10/2010] [Indexed: 01/28/2023] Open
Abstract
Background Peroxisome proliferator-activated receptor γ (PPAR γ) is a nuclear receptor whose activation has been shown to modulate macrophage and T cell-mediated inflammation. The objective of this study was to investigate the mechanisms by which the deletion of PPAR γ in T cells modulates immune cell distribution and colonic gene expression and the severity of experimental IBD. Methods PPAR γ flfl; CD4 Cre+ (CD4cre) or Cre- (WT) mice were challenged with 2.5% dextran sodium sulfate in their drinking water for 0, 2, or 7 days. Mice were scored on disease severity both clinically and histopathologically. Flow cytometry was used to assess lymphocyte and macrophage populations in the blood, spleen, and mesenteric lymph nodes (MLN). Global gene expression in colonic mucosa was profiled using Affymetrix microarrays. Results The deficiency of PPAR γ in T cells accelerated the onset of disease and body weight loss. Examination of colon histopathology revealed significantly greater epithelial erosion, leukocyte infiltration, and mucosal thickening in the CD4cre mice on day 7. CD4cre mice had more CD8+ T cells than WT mice and fewer CD4+FoxP3+ regulatory T cells (Treg) and IL10+CD4+ T cells in blood and MLN, respectively. Transcriptomic profiling revealed around 3000 genes being transcriptionally altered as a result of DSS challenge in CD4cre mice. These included up-regulated mRNA expression of adhesion molecules, proinflammatory cytokines interleukin-6 (IL-6) and IL-1β, and suppressor of cytokine signaling 3 (SOCS-3) on day 7. Gene set enrichment analysis (GSEA) showed that the ribosome and Krebs cycle pathways were downregulated while the apoptosis pathway was upregulated in colons of mice lacking PPAR γ in T cells. Conclusions The expression of PPAR γ in T cells is involved in preventing gut inflammation by regulating colonic expression of adhesion molecules and inflammatory mediators at later stages of disease while favoring the recruitment of Treg to the mucosal inductive sites.
Collapse
Affiliation(s)
- Amir J Guri
- Nutritional Immunology and Molecular Nutrition Laboratory, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | | | | | | | | |
Collapse
|