223 results on '"Yeaman MR"'
Search Results
2. PARIS and SPARTA: Finding the Achilles' Heel of SARS-CoV-2
- Author
-
Rasmussen, AL, Simon, V, Kota, V, Bloomquist, RF, Hanley, HB, Forgacs, D, Pahwa, S, Pallikkuth, S, Miller, LG, Schaenman, J, Yeaman, MR, Manthei, D, Wolf, J, Gaur, AH, Estepp, JH, Srivastava, K, Carreno, JM, Cuevas, F, Ellebedy, AH, Gordon, A, Valdez, R, Cobey, S, Reed, EF, Kolhe, R, Thomas, PG, Schultz-Cherry, S, Ross, TM, Krammer, F, Rasmussen, AL, Simon, V, Kota, V, Bloomquist, RF, Hanley, HB, Forgacs, D, Pahwa, S, Pallikkuth, S, Miller, LG, Schaenman, J, Yeaman, MR, Manthei, D, Wolf, J, Gaur, AH, Estepp, JH, Srivastava, K, Carreno, JM, Cuevas, F, Ellebedy, AH, Gordon, A, Valdez, R, Cobey, S, Reed, EF, Kolhe, R, Thomas, PG, Schultz-Cherry, S, Ross, TM, and Krammer, F
- Abstract
To understand reinfection rates and correlates of protection for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we established eight different longitudinal cohorts in 2020 under the umbrella of the PARIS (Protection Associated with Rapid Immunity to SARS-CoV-2)/SPARTA (SARS SeroPrevalence And Respiratory Tract Assessment) studies. Here, we describe the PARIS/SPARTA cohorts, the harmonized assays and analysis that are performed across the cohorts, as well as case definitions for SARS-CoV-2 infection and reinfection that have been established by the team of PARIS/SPARTA investigators. IMPORTANCE Determining reinfection rates and correlates of protection against SARS-CoV-2 infection induced by both natural infection and vaccination is of high significance for the prevention and control of coronavirus disease 2019 (COVID-19). Furthermore, understanding reinfections or infection after vaccination and the role immune escape plays in these scenarios will inform the need for updates of the current SARS-CoV-2 vaccines and help update guidelines suitable for the postpandemic world.
- Published
- 2022
3. Astrocytic outer retinal layer thinning is not a feature in AQP4-IgG seropositive neuromyelitis optica spectrum disorders
- Author
-
Altıntaş, Ayşe (ORCID 0000-0002-8524-5087 & YÖK ID 11611), Lu, A.; Zimmermann, HG.; Specovius, S.; Motamedi, S.; Chien, C.; Bereuter, C.; Lana-Peixoto, MA.; Fontenelle, MA.; Ashtari, F.; Kafieh, R.; Dehghani, A.; Pourazizi, M.; Pandit, L.; D Cunha, A.; Kim, HJ.; Hyun, JW.; Jung, SK.; Leocani, L.; Pisa, M.; Radaelli, M.; Siritho, S.; May, E.F.; Tongco, C.; De Sèze, J.; Senger, T.; Palace, J.; Roca-Fernández, A.; Leite, MI.; Sharma, SM.; Stiebel-Kalish, H.; Asgari, N.; Soelberg, K.K.; Martinez-Lapiscina, EH.; Havla, J.; Mao-Draayer, Y.; Rimler, Z.; Reid, A.; Marignier, R.; Cobo-Calvo, A.; Tanriverdi, U.; Yildirim, R.; Aktas, O.; Ringelstein, M.; Albrecht, P.; Tavares, IM.; Bichuetti, DB.; Jacob, A.; Huda, S.; Soto de Castillo, I.; Petzold, A.; Green, AJ.; Yeaman, MR.; Smith, TJ.; Cook, L.; Paul, F.; Brandt, AU.; Oertel, FC.; GJCF International Clinical Consortium for NMOSD., Koç University Research Center for Translational Medicine (KUTTAM) / Koç Üniversitesi Translasyonel Tıp Araştırma Merkezi (KUTTAM), School of Medicine, Altıntaş, Ayşe (ORCID 0000-0002-8524-5087 & YÖK ID 11611), Lu, A.; Zimmermann, HG.; Specovius, S.; Motamedi, S.; Chien, C.; Bereuter, C.; Lana-Peixoto, MA.; Fontenelle, MA.; Ashtari, F.; Kafieh, R.; Dehghani, A.; Pourazizi, M.; Pandit, L.; D Cunha, A.; Kim, HJ.; Hyun, JW.; Jung, SK.; Leocani, L.; Pisa, M.; Radaelli, M.; Siritho, S.; May, E.F.; Tongco, C.; De Sèze, J.; Senger, T.; Palace, J.; Roca-Fernández, A.; Leite, MI.; Sharma, SM.; Stiebel-Kalish, H.; Asgari, N.; Soelberg, K.K.; Martinez-Lapiscina, EH.; Havla, J.; Mao-Draayer, Y.; Rimler, Z.; Reid, A.; Marignier, R.; Cobo-Calvo, A.; Tanriverdi, U.; Yildirim, R.; Aktas, O.; Ringelstein, M.; Albrecht, P.; Tavares, IM.; Bichuetti, DB.; Jacob, A.; Huda, S.; Soto de Castillo, I.; Petzold, A.; Green, AJ.; Yeaman, MR.; Smith, TJ.; Cook, L.; Paul, F.; Brandt, AU.; Oertel, FC.; GJCF International Clinical Consortium for NMOSD., Koç University Research Center for Translational Medicine (KUTTAM) / Koç Üniversitesi Translasyonel Tıp Araştırma Merkezi (KUTTAM), and School of Medicine
- Abstract
Background: patients with anti-aquaporin-4 antibody seropositive (AQP4-IgG+) neuromyelitis optica spectrum disorders (NMOSDs) frequently suffer from optic neuritis (ON) leading to severe retinal neuroaxonal damage. Further, the relationship of this retinal damage to a primary astrocytopathy in NMOSD is uncertain. Primary astrocytopathy has been suggested to cause ON-independent retinal damage and contribute to changes particularly in the outer plexiform layer (OPL) and outer nuclear layer (ONL), as reported in some earlier studies. However, these were limited in their sample size and contradictory as to the localisation. This study assesses outer retinal layer changes using optical coherence tomography (OCT) in a multicentre cross-sectional cohort. Method: 197 patients who were AQP4-IgG+ and 32 myelin-oligodendrocyte-glycoprotein antibody seropositive (MOG-IgG+) patients were enrolled in this study along with 75 healthy controls. Participants underwent neurological examination and OCT with central postprocessing conducted at a single site. Results: no significant thinning of OPL (25.02 +/- 2.03 mu m) or ONL (61.63 +/- 7.04 mu m) were observed in patients who were AQP4-IgG+ compared with patients who were MOG-IgG+ with comparable neuroaxonal damage (OPL: 25.10 +/- 2.00 mu m; ONL: 64.71 +/- 7.87 mu m) or healthy controls (OPL: 24.58 +/- 1.64 mu m; ONL: 63.59 +/- 5.78 mu m). Eyes of patients who were AQP4-IgG+ (19.84 +/- 5.09 mu m, p=0.027) and MOG-IgG+ (19.82 +/- 4.78 mu m, p=0.004) with a history of ON showed parafoveal OPL thinning compared with healthy controls (20.99 +/- 5.14 mu m); this was not observed elsewhere. Conclusion: the results suggest that outer retinal layer loss is not a consistent component of retinal astrocytic damage in AQP4-IgG+ NMOSD. Longitudinal studies are necessary to determine if OPL and ONL are damaged in late disease due to retrograde trans-synaptic axonal degeneration and whether outer retinal dysfunction occurs despite any measurable st, Guthy Jackson Charitable Foundation (GJCF); German Research Foundation (DFG)
- Published
- 2021
4. New approaches to the prevention and treatment of severe S. aureus infections
- Author
-
Bayer As, Yeaman Mr, and Yan Q. Xiong
- Subjects
business.industry ,medicine.drug_class ,Osteomyelitis ,Antibiotics ,medicine.disease ,Antimicrobial ,medicine.disease_cause ,Microbiology ,Sepsis ,Staphylococcus aureus ,Infective endocarditis ,medicine ,Vancomycin ,Septic arthritis ,business ,medicine.drug - Abstract
Staphylococcus aureus is a virulent pathogen that is currently a major cause of community-acquired infections, as well as infections in hospitalized patients. Morbidity and mortality due to S. aureus infections, such as sepsis, osteomyelitis, septic arthritis and infective endocarditis, remain high despite the use of newer antibiotics. Of major concern, methicillin resistance in S. aureus isolates has increased dramatically worldwide, especially among nosocomial isolates; this phenotype may be associated with resistance to other antistaphylococcal compounds, including vancomycin. This increase in prevalence of multiantibiotic resistance in S. aureus is a major public health concern. Currently, there is an intense focus on the development of novel vaccines for the prevention of S. aureus infections in high-risk populations and on new antimicrobial classes for the therapy of established S. aureus infections.
- Published
- 2003
5. Linezolid: A new antibiotic
- Author
-
Bayer As, Yan Q. Xiong, and Yeaman Mr
- Subjects
Pharmacology ,biology ,business.industry ,medicine.drug_class ,Antibiotics ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,bacterial infections and mycoses ,Antimicrobial ,medicine.disease ,biology.organism_classification ,Peptostreptococcus ,Microbiology ,chemistry.chemical_compound ,chemistry ,Linezolid ,bacteria ,Medicine ,Endocarditis ,Vancomycin ,Pharmacology (medical) ,business ,Meningitis ,Antibacterial agent ,medicine.drug - Abstract
The U.S. Food and Drug Administration recently approved linezolid for the treatment of patients with methicillin-resistant staphylococcal and vancomycin-resistant enterococcal infections. This oxazolidinone antibacterial agent represents the first approved antibiotic of a new structural class in 35 years. Linezolid is a synthetic compound that acts by inhibiting the initiation complex formation in bacterial protein synthesis, a mechanism of action distinct from other commercially available antibiotics. Thus, cross-resistance between linezolid and other current antimicrobial agents has not been demonstrated to date. Linezolid has a wide spectrum of in vitro activity against Gram-positive organisms, including methicillin-resistant staphylococci, penicillin-resistant pneumococci and vancomycin-resistant enterococci. Some anaerobes, such as Clostridium spp., Peptostreptococcus spp. and Prevotella spp. are also susceptible to linezolid. In addition, linezolid has exhibited good efficacy in experimental animal models of acute otitis media, endocarditis and meningitis due to many common aerobic Gram-positive bacteria. In clinical trials involving hospitalized patients with skin/soft tissue infections, community-acquired pneumonia and serious Gram-positive bacterial infections, linezolid appeared to be an effective treatment option, comparable in efficacy to vancomycin.
- Published
- 2000
- Full Text
- View/download PDF
6. Regulation of Staphylococcus aureus alpha-toxin gene (hla) expression by agr, sarA, and sae in vitro and in experimental infective endocarditis.
- Author
-
Xiong YQ, Willard J, Yeaman MR, Cheung AL, and Bayer AS
- Abstract
BACKGROUND: The Staphylococcus aureus global regulators--agr, sarA, and sae--coordinately control alpha-toxin gene (hla) expression in vitro. However, their relative in vivo contributions to hla expression, particularly in endovascular infections, have not been defined. METHODS: A plasmid-based hla-promoter:green fluorescent protein reporter system was constructed in 2 genetically related S. aureus strains: RN6390 (a natural sigma factor B [sigB]-deficient mutant), SH1000 (a sigB-repaired variant of RN6390 lineage), and their respective agr, sarA, agr/sarA, and sae mutants. These strain sets were used to quantify hla expression in vitro and in an experimental infective endocarditis (IE) model using flow cytometry. RESULTS: In vitro, hla expression was positively modulated by all 3 regulons (sae > agr/sarA > agr and sarA) in both RN6390 and SH1000 backgrounds. In the IE model, hla expression in cardiac vegetations was lower in all single mutants than in the respective parental strains (P<.05 for sae mutant) but was maintained at near-parental levels in the agr/sarA double mutant in both backgrounds. A similar finding was also observed in kidneys and spleens. CONCLUSIONS: These results indicate that, although several distinct regulatory circuits can affect hla expression in vitro and in vivo, sae appears to play a crucial role in this context. Copyright © 2006 Infectious Diseases Society of America [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
7. Astrocytic outer retinal layer thinning is not a feature in AQP4-IgG seropositive neuromyelitis optica spectrum disorders
- Author
-
Maria Isabel Leite, Letizia Leocani, Jérôme De Seze, Denis Bernardi Bichuetti, Ibis Soto de Castillo, Sasitorn Siritho, Alvaro Cobo-Calvo, Elena H. Martinez-Lapiscina, Anitha D'Cunha, Adriana Roca-Fernandez, Jacqueline Palace, Orhan Aktas, Marco Aurélio Lana-Peixoto, Marco Pisa, Michael R. Yeaman, Anu Jacob, Mariana Andrade Fontenelle, Friedemann Paul, Ari J. Green, Yang Mao-Draayer, Eugene F May, Claudia Chien, Alireza Dehghani, Ivan Maynart Tavares, Lawrence J. Cook, Angelo Lu, Su-Kyung Jung, Rengin Yildirim, Ayse Altintas, Mohsen Pourazizi, Ho Jin Kim, Rahele Kafieh, Caryl Tongco, Lekha Pandit, Hadas Stiebel-Kalish, Romain Marignier, Fereshteh Ashtari, Hanna Zimmermann, Joachim Havla, M. Radaelli, Svenja Specovius, Frederike C. Oertel, Kerstin Soelberg, Srilakshmi M Sharma, Axel Petzold, Seyedamirhosein Motamedi, Zoe Rimler, Saif Huda, Philipp Albrecht, Alexander U. Brandt, Jae-Won Hyun, Allyson Reid, Marius Ringelstein, Uygur Tanriverdi, Terry J. Smith, Thomas Senger, Nasrin Asgari, Charlotte Bereuter, Altıntaş, Ayşe (ORCID 0000-0002-8524-5087 & YÖK ID 11611), Lu, A., Zimmermann, HG., Specovius, S., Motamedi, S., Chien, C., Bereuter, C., Lana-Peixoto, MA., Fontenelle, MA., Ashtari, F., Kafieh, R., Dehghani, A., Pourazizi, M., Pandit, L., D Cunha, A., Kim, HJ., Hyun, JW., Jung, SK., Leocani, L., Pisa, M., Radaelli, M., Siritho, S., May, E.F., Tongco, C., De Sèze, J., Senger, T., Palace, J., Roca-Fernández, A., Leite, MI., Sharma, SM., Stiebel-Kalish, H., Asgari, N., Soelberg, K.K., Martinez-Lapiscina, EH., Havla, J., Mao-Draayer, Y., Rimler, Z., Reid, A., Marignier, R., Cobo-Calvo, A., Tanriverdi, U., Yildirim, R., Aktas, O., Ringelstein, M., Albrecht, P., Tavares, IM., Bichuetti, DB., Jacob, A., Huda, S., Soto de Castillo, I., Petzold, A., Green, AJ., Yeaman, MR., Smith, TJ., Cook, L., Paul, F., Brandt, AU., Oertel, FC., GJCF International Clinical Consortium for NMOSD., Koç University Research Center for Translational Medicine (KUTTAM) / Koç Üniversitesi Translasyonel Tıp Araştırma Merkezi (KUTTAM), School of Medicine, Neurology, Ophthalmology, APH - Mental Health, APH - Methodology, and Amsterdam Neuroscience - Neuroinfection & -inflammation
- Subjects
Adult ,Male ,medicine.medical_specialty ,genetic structures ,Vision ,Clinical neurology ,Ophthalmology ,Outer plexiform layer ,Retina ,chemistry.chemical_compound ,medicine ,Humans ,In patient ,Optic neuritis ,Outer nuclear layer ,Autoantibodies ,Aquaporin 4 ,business.industry ,Neuromyelitis Optica ,Retinal ,Middle Aged ,medicine.disease ,eye diseases ,Neurosciences and neurology ,Psychiatry ,Surgery ,Psychiatry and Mental health ,medicine.anatomical_structure ,Retinal dysfunction ,Cross-Sectional Studies ,chemistry ,Neuromyelitis Optica Spectrum Disorders ,Astrocytes ,Female ,Neurology (clinical) ,sense organs ,business ,Function and Dysfunction of the Nervous System ,Axonal degeneration ,Tomography, Optical Coherence - Abstract
Background: patients with anti-aquaporin-4 antibody seropositive (AQP4-IgG+) neuromyelitis optica spectrum disorders (NMOSDs) frequently suffer from optic neuritis (ON) leading to severe retinal neuroaxonal damage. Further, the relationship of this retinal damage to a primary astrocytopathy in NMOSD is uncertain. Primary astrocytopathy has been suggested to cause ON-independent retinal damage and contribute to changes particularly in the outer plexiform layer (OPL) and outer nuclear layer (ONL), as reported in some earlier studies. However, these were limited in their sample size and contradictory as to the localisation. This study assesses outer retinal layer changes using optical coherence tomography (OCT) in a multicentre cross-sectional cohort. Method: 197 patients who were AQP4-IgG+ and 32 myelin-oligodendrocyte-glycoprotein antibody seropositive (MOG-IgG+) patients were enrolled in this study along with 75 healthy controls. Participants underwent neurological examination and OCT with central postprocessing conducted at a single site. Results: no significant thinning of OPL (25.02 +/- 2.03 mu m) or ONL (61.63 +/- 7.04 mu m) were observed in patients who were AQP4-IgG+ compared with patients who were MOG-IgG+ with comparable neuroaxonal damage (OPL: 25.10 +/- 2.00 mu m; ONL: 64.71 +/- 7.87 mu m) or healthy controls (OPL: 24.58 +/- 1.64 mu m; ONL: 63.59 +/- 5.78 mu m). Eyes of patients who were AQP4-IgG+ (19.84 +/- 5.09 mu m, p=0.027) and MOG-IgG+ (19.82 +/- 4.78 mu m, p=0.004) with a history of ON showed parafoveal OPL thinning compared with healthy controls (20.99 +/- 5.14 mu m); this was not observed elsewhere. Conclusion: the results suggest that outer retinal layer loss is not a consistent component of retinal astrocytic damage in AQP4-IgG+ NMOSD. Longitudinal studies are necessary to determine if OPL and ONL are damaged in late disease due to retrograde trans-synaptic axonal degeneration and whether outer retinal dysfunction occurs despite any measurable structural correlates., Guthy Jackson Charitable Foundation (GJCF); German Research Foundation (DFG)
- Published
- 2021
- Full Text
- View/download PDF
8. Application and interpretation of core elements of the 2015 NMOSD diagnostic criteria in routine clinical practice.
- Author
-
Carnero Contentti E, Rojas JI, Alonso R, Yeaman MR, and Weinshenker BG
- Subjects
- Humans, Female, Male, Cross-Sectional Studies, Middle Aged, Adult, Surveys and Questionnaires, Neuromyelitis Optica diagnosis, Magnetic Resonance Imaging, Neurologists
- Abstract
Background: We evaluated comprehension and application of the 2015 neuromyelitis optica spectrum disorder (NMOSD) criteria core elements by neurologists in Latin America (LATAM) who routinely diagnose and care for NMOSD patients by (i) identifying typical/suggestive NMOSD syndromes, (ii) detecting typical MRI NMOSD lesions and meeting MRI dissemination in space (DIS) criteria, and (iii) evaluating historical symptoms suggestive of NMOSD., Methods: We conducted an anonymous, voluntary, self-administered web- and case-based survey cross-sectional study from October 2023 to January 2024 of neurologists identified through the LACTRIMS database. Questions were presented first through iterative clinical cases or imaging, followed by questions directly evaluating comprehension of definitions. "Correct" responses were based on the 2015 criteria and adjudicated by the consensus of the experts leading the project., Results: A total of 106 neurologists (60.3% female; mean age: 46.6 ± 12.5 years) were included. Between 10.4% and 49.1% of neurologists inaccurately identified clinical or paraclinical aspects for DIS and 32.1% accurately identified the three non-cardinal (brainstem, diencephalic, and cerebral) syndromes for seronegative patients. Between 35.8% and 64.1% of neurologists identified the "optimal timing" of AQP4-IgG testing (e.g., during an attack or before receiving immunosuppressant treatments, among others); 56.6% considered live cell-based assay as the gold standard method for serological testing. Most neurologists accurately identified typical NMOSD MRI lesions, but periventricular, juxtacortical/cortical, fluffy infratentorial, corticospinal tract, and hypothalamic lesions were frequently misidentified., Conclusion: Clinical scenarios were identified where the 2015 NMOSD criteria were susceptible to misinterpretation and misapplication by expert neurologists in LATAM. Implementing collaborative educational initiatives could improve NMOSD diagnosis and raise patient care standards., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Carnero Contentti, Rojas, Alonso, Yeaman and Weinshenker.)
- Published
- 2024
- Full Text
- View/download PDF
9. Aquaporin-4 Immunoglobulin G-seropositive Neuromyelitis Optica Spectrum Disorder MRI Characteristics: Data Analysis from the International Real-World PAMRINO Study Cohort.
- Author
-
Chien C, Cruz E Silva V, Geiter E, Meier D, Zimmermann H, Bichuetti DB, Idagawa MI, Altintas A, Tanriverdi U, Siritho S, Pandit L, Dcunha A, Sá MJ, Figueiredo R, Qian P, Tongco C, Lotan I, Khasminsky V, Hellmann MA, Stiebel-Kalish H, Rotstein DL, Waxman L, Ontaneda D, Nakamura K, Abboud H, Subei MO, Mao-Draayer Y, Havla J, Asgari N, Skejø PB, Kister I, Ringelstein M, Broadley S, Arnett S, Marron B, Jolley AM, Wunderlich M, Green S, Cook LJ, Yeaman MR, Smith TJ, Brandt AU, Wuerfel J, and Paul F
- Subjects
- Humans, Female, Male, Adult, Retrospective Studies, Middle Aged, Cross-Sectional Studies, Aged, Young Adult, Adolescent, Cohort Studies, Neuromyelitis Optica diagnostic imaging, Neuromyelitis Optica immunology, Magnetic Resonance Imaging methods, Aquaporin 4 immunology, Immunoglobulin G blood
- Abstract
Background Patients with neuromyelitis optica spectrum disorder (NMOSD) are often seropositive for antibodies against aquaporin-4 (AQP4). The importance of MRI monitoring in this disease requires evaluation. Purpose To profile MRI features from a large international cohort with AQP4 immunoglobulin G (IgG)-seropositive NMOSD (from the Parallel MRI in NMOSD [PAMRINO] study) and to evaluate and confirm existing knowledge regarding the incidence, location, and longitudinal development of characteristic lesions in the central nervous system associated with AQP4-IgG-seropositive NMOSD. Materials and Methods In this retrospective study (from August 2016 to January 2019), MRI and clinical data were collected from 17 NMOSD expert sites in 11 countries across four continents. Clinical features and lesions identified at cross-sectional and longitudinal MRI were assessed. No formal statistical tests were used to compare observations; however, means, SDs, and 95% CIs are reported when evaluating lesion frequencies. Results Available T1-weighted and T2-weighted MRI scans in patients with AQP4-IgG-seropositive NMOSD ( n = 525) were read. Among the 525 patients, 320 underwent cerebral MRI examinations with T2-weighted hyperintense cerebral (264 of 320; 82.5%), cerebellar (44 of 320; 13.8%), and brainstem (158 of 321 [49.2%], including one lesion observed at cervical spinal cord [SC] MRI) lesions. Lesions in the optic nerves, analyzed from 152 MRI examinations, were mainly found in the central (81 of 92; 88%) and posterior (79 of 92; 86%) sections (bilaterally in 39 of 92; 42%). Longitudinally extensive transverse myelitis was the predominant SC lesion pattern (upper compartment from 322 MRI examinations, 133 of 210 [63.3%]; and lower compartment from 301 MRI examinations, 149 of 212 [70.3%]). However, nonlongitudinal extensive transverse myelitis lesions were also observed frequently (105 of 210; 50.0%) in the cervical SC. Clinical data ( n = 349; mean age, 44 years ± 14 [SD]; 202 female patients) and acute lesions at contrast-enhanced (CE) MRI ( n = 58, performed within 30 days of the last attack) were evaluated. CE lesions were detected in the cerebrum (eight of 13; 62%), optic nerves (14 of 19; 74%), or chiasm (three of four; 75%) within 15 days of any relapse. In the upper SC (29 of 44; 66%), CE lesions were frequently observed up to 20 days after a clinical myelitis event. Conclusion A high incidence of abnormal brain MRI examinations and nonlongitudinal extensive SC lesions was found in patients in PAMRINO with AQP4-IgG-seropositive NMOSD. © RSNA, 2024 Supplemental material is available for this article.
- Published
- 2024
- Full Text
- View/download PDF
10. Diagnostic Value of Inter-Eye Difference Metrics on OCT for Myelin Oligodendrocyte Glycoprotein Antibody-Associated Optic Neuritis.
- Author
-
Volpe G, Jurkute N, Girafa G, Zimmermann HG, Motamedi S, Bereuter C, Pandit L, D'Cunha A, Yeaman MR, Smith TJ, Cook LJ, Brandt AU, Paul F, Petzold A, and Oertel FC
- Subjects
- Humans, Female, Male, Adult, Middle Aged, Sensitivity and Specificity, Young Adult, Myelin-Oligodendrocyte Glycoprotein immunology, Optic Neuritis immunology, Optic Neuritis diagnosis, Optic Neuritis diagnostic imaging, Tomography, Optical Coherence, Autoantibodies blood
- Abstract
Background and Objectives: The 2022 International Consortium for Optic Neuritis diagnostic criteria for optic neuritis (ON) include optical coherence tomography (OCT). The diagnostic value of intereye difference (IED) metrics is high for ON in patients with multiple sclerosis and aquaporin-4 antibody seropositive neuromyelitis optica spectrum disorders, but unknown in myelin oligodendrocyte glycoprotein antibody-associated ON (MOG-ON)., Methods: A multicenter validation study was conducted on the published IED cutoff values (>4% or >4 μm in the macular ganglion cell and inner plexiform layer [mGCIP] or >5% or >5 μm in the peripapillary retinal nerve fiber layer [pRNFL]) in individuals with MOG-ON and age-matched and sex-matched healthy controls (HCs). Structural data were acquired with Spectralis spectral-domain OCT >6 months after ON. We calculated sensitivity, specificity, and receiver operating characteristics for both intereye percentage (IEPD) and absolute difference (IEAD)., Results: A total of 66 individuals were included (MOG-ON N = 33; HCs N = 33). ON was unilateral in 20 and bilateral in 13 subjects. In the pooled analysis, the mGCIP IEPD was most sensitive (92%), followed by the mGCIP IEAD (88%) and pRNFL (84%). The same pattern was found for the specificity (mGCIP IEPD 82%, IEAD 82%; pRNFL IEPD 82%, IEAD 79%).In subgroup analyses, the diagnostic sensitivity was higher in subjects with unilateral ON (>99% for all metrics) compared with bilateral ON (61%-78%)., Discussion: In individuals with MOG-ON, the diagnostic accuracy of OCT-based IED metrics for ON was high, especially of mGCIP IEPD., Classification of Evidence: This study provides Class III evidence that the intereye difference on OCT can distinguish between those with MOG and normal controls.
- Published
- 2024
- Full Text
- View/download PDF
11. Assessment of disability and disease burden in neuromyelitis optica spectrum disorders in the CIRCLES Cohort.
- Author
-
Gholizadeh S, Exuzides A, Sinnott J, Palmer C, Waltz M, Rose JW, Jolley AM, Behne JM, Behne MK, Blaschke TF, Smith TJ, Lewis KE, Cook LJ, and Yeaman MR
- Subjects
- Humans, Female, Male, Middle Aged, Adult, Cohort Studies, Disabled Persons, Cost of Illness, Aquaporin 4 immunology, Immunoglobulin G blood, Severity of Illness Index, Longitudinal Studies, Neuromyelitis Optica immunology, Disability Evaluation
- Abstract
Neuromyelitis optica spectrum disorders (NMOSD) comprise autoimmune diseases imposing substantial disability. We compared an NMOSD-targeted disability assessment of mobility, vision, and self-care domains (individually and composite) with the multiple sclerosis-targeted Expanded Disability Status Scale (EDSS) to assess NMOSD disease burden. An overall cohort (n = 505) and a subset of these patients with an enriched dataset (n = 198) were analyzed from the CIRCLES longitudinal, observational database of patients with AQP4-IgG-seropositive or -seronegative NMOSD in North America. Multinomial modeling was used to identify temporal correlates of disability improvement, stability, and worsening. Prior on-study relapse correlated with worsening mobility (OR, 3.08; 95% CI: 1.61-5.90), vision (OR, 3.99; 95% CI: 2.03-7.86), self-care disability (OR, 1.90; 95% CI: 1.07-3.38), and mean composite index disability (OR, 4.20; 95% CI: 1.71-10.34). Higher vision disability was associated with Black race, shorter time on-study, and AQP4-IgG-seropositive status in patients ≥ 18 years (p < 0.05). Disease onset phenotype and sex correlated with pain interference (p < 0.05). These correlates of NMOSD disability were undetected by EDSS. The CIRCLES real-world experience supports the need for NMOSD-specific disability assessment to improve recognition of disease burden, facilitate proactive clinical management, offer insights into resilience, and inform clinical trial design., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
12. Sulfated glycosaminoglycans are host epithelial cell targets of the Candida albicans toxin candidalysin.
- Author
-
Lin J, Miao J, Schaefer KG, Russell CM, Pyron RJ, Zhang F, Phan QT, Solis NV, Liu H, Tashiro M, Dordick JS, Linhardt RJ, Yeaman MR, King GM, Barrera FN, Peters BM, and Filler SG
- Subjects
- Animals, Humans, Mice, Female, Cell Line, Virulence Factors metabolism, Virulence Factors genetics, Cytokines metabolism, Candida albicans drug effects, Candida albicans metabolism, Candida albicans genetics, Epithelial Cells microbiology, Epithelial Cells metabolism, Epithelial Cells drug effects, Fungal Proteins metabolism, Fungal Proteins genetics, Dextran Sulfate, Glycosaminoglycans metabolism, Candidiasis, Vulvovaginal microbiology, Candidiasis, Vulvovaginal drug therapy
- Abstract
Candidalysin, a cytolytic peptide produced by the fungal pathogen Candida albicans, is a key virulence factor. However, its host cell targets remain elusive. Here we performed a genome-wide loss-of-function CRISPR screen in the TR146 human oral epithelial cell line and identified that disruption of genes (XYLT2, B3GALT6 and B3GAT3) in glycosaminoglycan (GAG) biosynthesis conferred resistance to damage induced by candidalysin and live C. albicans. Surface plasmon resonance and atomic force and electron microscopy indicated that candidalysin binds to sulfated GAGs, facilitating its enrichment on the host cell surface. Adding exogenous sulfated GAGs or the analogue dextran sulfate protected cells against candidalysin-induced damage. Dextran sulfate also inhibited C. albicans invasion and fungal-induced epithelial cell cytokine production. In mice with vulvovaginal candidiasis, topical dextran sulfate administration reduced intravaginal tissue damage and inflammation. Collectively, sulfated GAGs are epithelial cell targets of candidalysin and can be used therapeutically to protect cells from candidalysin-induced damage., (© 2024. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
13. The Archetypal Gamma-Core Motif of Antimicrobial Cys-Rich Peptides Inhibits H + -ATPases in Target Pathogens.
- Author
-
Andrés MT, Yount NY, Acosta-Zaldívar M, Yeaman MR, and Fierro JF
- Subjects
- Humans, Antimicrobial Peptides pharmacology, Antimicrobial Peptides chemistry, Lactoferrin pharmacology, Lactoferrin chemistry, Anti-Infective Agents pharmacology, Anti-Infective Agents chemistry, Cysteine metabolism, Cysteine chemistry, Candida albicans drug effects, Cell Membrane metabolism, Cell Membrane drug effects, Proton-Translocating ATPases metabolism, Proton-Translocating ATPases antagonists & inhibitors, Amino Acid Motifs
- Abstract
Human lactoferrin (hLf) is an innate host defense protein that inhibits microbial H
+ -ATPases. This protein includes an ancestral structural motif (i.e., γ-core motif) intimately associated with the antimicrobial activity of many natural Cys-rich peptides. Peptides containing a complete γ-core motif from hLf or other phylogenetically diverse antimicrobial peptides (i.e., afnA, SolyC, PA1b, Pv D1 , thanatin) showed microbicidal activity with similar features to those previously reported for hLf and defensins. Common mechanistic characteristics included (1) cell death independent of plasma membrane (PM) lysis, (2) loss of intracellular K+ (mediated by Tok1p K+ channels in yeast), (3) inhibition of microbicidal activity by high extracellular K+ , (4) influence of cellular respiration on microbicidal activity, (5) involvement of mitochondrial ATP synthase in yeast cell death processes, and (6) increment of intracellular ATP. Similar features were also observed with the BM2 peptide, a fungal PM H+ -ATPase inhibitor. Collectively, these findings suggest host defense peptides containing a homologous γ-core motif inhibit PM H+ -ATPases. Based on this discovery, we propose that the γ-core motif is an archetypal effector involved in the inhibition of PM H+ -ATPases across kingdoms of life and contributes to the in vitro microbicidal activity of Cys-rich antimicrobial peptides.- Published
- 2024
- Full Text
- View/download PDF
14. Retinal Changes in Double-Antibody Seronegative Neuromyelitis Optica Spectrum Disorders.
- Author
-
Oertel FC, Zimmermann HG, Motamedi S, Bereuter C, Manthey LM, Ashtari F, Kafieh R, Dehghani A, Pourazizi M, Pandit L, D'Cunha A, Aktas O, Albrecht P, Ringelstein M, Martinez-Lapiscina EH, Sanchez Dalmau BF, Villoslada P, Asgari N, Marignier R, Cobo-Calvo A, Leocani L, Pisa M, Radaelli M, Palace J, Roca-Fernandez A, Leite MIS, Sharma S, De Seze J, Senger T, Yeaman MR, Smith TJ, Cook LJ, Brandt AU, and Paul F
- Subjects
- Humans, Female, Male, Adult, Cross-Sectional Studies, Middle Aged, Retrospective Studies, Autoantibodies blood, Retina diagnostic imaging, Retina pathology, Retina immunology, Neuromyelitis Optica diagnostic imaging, Neuromyelitis Optica immunology, Neuromyelitis Optica blood, Tomography, Optical Coherence, Aquaporin 4 immunology
- Abstract
Background and Objectives: To systematically describe the clinical picture of double-antibody seronegative neuromyelitis optica spectrum disorders (DN-NMOSD) with specific emphasis on retinal involvement., Methods: Cross-sectional data of 25 people with DN-NMOSD (48 eyes) with and without a history of optic neuritis (ON) were included in this study along with data from 25 people with aquaporin-4 antibody seropositive neuromyelitis optica spectrum disorder (AQP4-NMOSD, 46 eyes) and from 25 healthy controls (HCs, 49 eyes) for comparison. All groups were matched for age and sex and included from the collaborative retrospective study of retinal optical coherence tomography (OCT) in neuromyelitis optica (CROCTINO). Participants underwent OCT with central postprocessing and local neurologic examination and antibody testing. Retinal neurodegeneration was quantified as peripapillary retinal nerve fiber layer thickness (pRNFL) and combined ganglion cell and inner plexiform layer thickness (GCIPL)., Results: This DN-NMOSD cohort had a history of [median (inter-quartile range)] 6 (5; 9) attacks within their 5 ± 4 years since onset. Myelitis and ON were the most common attack types. In DN-NMOSD eyes after ON, pRNFL ( p < 0.001) and GCIPL ( p = 0.023) were thinner compared with eyes of HCs. Even after only one ON episode, DN-NMOSD eyes already had considerable neuroaxonal loss compared with HCs. In DN-NMOSD eyes without a history of ON, pRNFL ( p = 0.027) and GCIPL ( p = 0.022) were also reduced compared with eyes of HCs. However, there was no difference in pRNFL and GCIPL between DN-NMOSD and AQP4-NMOSD for the whole group and for subsets with a history of ON and without a history of ON-as well as between variances of retinal layer thicknesses., Discussion: DN-NMOSD is characterized by severe retinal damage after ON and attack-independent retinal neurodegeneration. Most of the damage occurs during the first ON episode, which highlights the need for better diagnostic markers in DN-NMOSD to facilitate an earlier diagnosis as well as for effective and early treatments. In this study, people with DN-NMOSD presented with homogeneous clinical and imaging findings potentially suggesting a common retinal pathology in these patients.
- Published
- 2024
- Full Text
- View/download PDF
15. Sex ratio and age of onset in AQP4 antibody-associated NMOSD: a review and meta-analysis.
- Author
-
Arnett S, Chew SH, Leitner U, Hor JY, Paul F, Yeaman MR, Levy M, Weinshenker BG, Banwell BL, Fujihara K, Abboud H, Dujmovic Basuroski I, Arrambide G, Neubrand VE, Quan C, Melamed E, Palace J, Sun J, Asgari N, and Broadley SA
- Subjects
- Humans, Female, Male, Sex Ratio, Neuromyelitis Optica immunology, Neuromyelitis Optica blood, Aquaporin 4 immunology, Age of Onset, Autoantibodies blood
- Abstract
Background: Aquaporin-4 (AQP4) antibody-associated neuromyelitis optica spectrum disorder (NMOSD) is an antibody-mediated inflammatory disease of the central nervous system. We have undertaken a systematic review and meta-analysis to ascertain the sex ratio and mean age of onset for AQP4 antibody associated NMOSD. We have also explored factors that impact on these demographic data., Methods: A systematic search of databases was conducted according to the PRISMA guidelines. Articles reporting sex distribution and age of onset for AQP4 antibody-associated NMSOD were reviewed. An initially inclusive approach involving exploration with regression meta-analysis was followed by an analysis of just AQP4 antibody positive cases., Results: A total of 528 articles were screened to yield 89 articles covering 19,415 individuals from 88 population samples. The female:male sex ratio was significantly influenced by the proportion of AQP4 antibody positive cases in the samples studied (p < 0.001). For AQP4 antibody-positive cases the overall estimate of the sex ratio was 8.89 (95% CI 7.78-10.15). For paediatric populations the estimate was 5.68 (95% CI 4.01-8.03) and for late-onset cases, it was 5.48 (95% CI 4.10-7.33). The mean age of onset was significantly associated with the mean life expectancy of the population sampled (p < 0.001). The mean age of onset for AQP4 antibody-positive cases in long-lived populations was 41.7 years versus 33.3 years in the remainder., Conclusions: The female:male sex ratio and the mean age of onset of AQP4 antibody-associated NMOSD are significantly higher than MS. The sex ratio increases with the proportion of cases that are positive for AQP4 antibodies and the mean age of onset increases with population life expectancy., (© 2024. Crown.)
- Published
- 2024
- Full Text
- View/download PDF
16. The Purine Biosynthesis Repressor, PurR, Contributes to Vancomycin Susceptibility of Methicillin-resistant Staphylococcus aureus in Experimental Endocarditis.
- Author
-
Xiong YQ, Li Y, Goncheva MI, Elsayed AM, Zhu F, Li L, Abdelhady W, Flannagan RS, Yeaman MR, Bayer AS, and Heinrichs DE
- Subjects
- Animals, Virulence Factors genetics, Virulence Factors metabolism, Mice, Gene Expression Regulation, Bacterial, Disease Models, Animal, Microbial Sensitivity Tests, Humans, Methicillin-Resistant Staphylococcus aureus drug effects, Methicillin-Resistant Staphylococcus aureus genetics, Methicillin-Resistant Staphylococcus aureus metabolism, Bacterial Proteins genetics, Bacterial Proteins metabolism, Staphylococcal Infections microbiology, Staphylococcal Infections drug therapy, Purines biosynthesis, Anti-Bacterial Agents pharmacology, Vancomycin pharmacology, Repressor Proteins genetics, Repressor Proteins metabolism, Endocarditis, Bacterial microbiology, Endocarditis, Bacterial drug therapy
- Abstract
Background: Staphylococcus aureus is the most common cause of life-threatening endovascular infections, including infective endocarditis (IE). These infections, especially when caused by methicillin-resistant strains (MRSA), feature limited therapeutic options and high morbidity and mortality rates., Methods: Herein, we investigated the role of the purine biosynthesis repressor, PurR, in virulence factor expression and vancomycin (VAN) treatment outcomes in experimental IE due to MRSA., Results: The PurR-mediated repression of purine biosynthesis was confirmed by enhanced purF expression and production of an intermediate purine metabolite in purR mutant strain. In addition, enhanced expression of the transcriptional regulators, sigB and sarA, and their key downstream virulence genes (eg, fnbA, and hla) was demonstrated in the purR mutant in vitro and within infected cardiac vegetations. Furthermore, purR deficiency enhanced fnbA/fnbB transcription, translating to increased fibronectin adhesion versus the wild type and purR-complemented strains. Notably, the purR mutant was refractory to significant reduction in target tissues MRSA burden following VAN treatment in the IE model., Conclusions: These findings suggest that the purine biosynthetic pathway intersects the coordination of virulence factor expression and in vivo persistence during VAN treatment, and may represent an avenue for novel antimicrobial development targeting MRSA., Competing Interests: Potential conflicts of interest. All authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2024. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
17. Integrated transcriptomic analysis reveals immune signatures distinguishing persistent versus resolving outcomes in MRSA bacteremia.
- Author
-
Parmar R, Pickering H, Ahn R, Rossetti M, Gjertson DW, Ruffin F, Chan LC, Fowler VG Jr, Yeaman MR, and Reed EF
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Interleukin-10 genetics, Interleukin-10 blood, DNA Methyltransferase 3A, Anti-Bacterial Agents therapeutic use, Adult, Bacteremia diagnosis, Bacteremia immunology, Bacteremia genetics, Bacteremia microbiology, Methicillin-Resistant Staphylococcus aureus, Staphylococcal Infections immunology, Staphylococcal Infections genetics, Staphylococcal Infections diagnosis, Staphylococcal Infections microbiology, Gene Expression Profiling, Transcriptome
- Abstract
Introduction: Staphylococcus aureus bacteremia (SAB) is a life-threatening infection particularly involving methicillin-resistant S. aureus (MRSA). In contrast to resolving MRSA bacteremia (RB), persistent MRSA bacteremia (PB) blood cultures remain positive despite appropriate antibiotic treatment. Host immune responses distinguishing PB vs. RB outcomes are poorly understood. Here, integrated transcriptomic, IL-10 cytokine levels, and genomic analyses sought to identify signatures differentiating PB vs. RB outcomes., Methods: Whole-blood transcriptomes of propensity-matched PB (n=28) versus RB (n=30) patients treated with vancomycin were compared in one independent training patient cohort. Gene expression (GE) modules were analyzed and prioritized relative to host IL-10 cytokine levels and DNA methyltransferase-3A ( DNMT3A ) genotype., Results: Differential expression of T and B lymphocyte gene expression early in MRSA bacteremia discriminated RB from PB outcomes. Significant increases in effector T and B cell signaling pathways correlated with RB, lower IL-10 cytokine levels and DNMT3A heterozygous A/C genotype. Importantly, a second PB and RB patient cohort analyzed in a masked manner demonstrated high predictive accuracy of differential signatures., Discussion: Collectively, the present findings indicate that human PB involves dysregulated immunity characterized by impaired T and B cell responses associated with excessive IL-10 expression in context of the DNMT3A A/A genotype. These findings reveal distinct immunologic programs in PB vs. RB outcomes, enable future studies to define mechanisms by which host and/or pathogen drive differential signatures and may accelerate prediction of PB outcomes. Such prognostic assessment of host risk could significantly enhance early anti-infective interventions to avert PB and improve patient outcomes., Competing Interests: VGF reports Grant/Research Support: MedImmune, Cerexa/Forest/Actavis/Allergan, Pfizer, Advanced Liquid Logics, Theravance, Novartis, Cubist/Merck; Medical Biosurfaces; Locus; Affinergy; Contrafect; Karius; Genentech, Regeneron, BasileaPaid Consultant: Pfizer, Novartis Galderma, Novadigm, Durata, Debiopharm, Genentech, Achaogen, Affinium, Medicines Co., Cerexa, Tetraphase, Trius, MedImmune, Bayer, Theravance, Cubist, Basilea, Affinergy, Janssen, xBiotech, Contrafect, Regeneron, Basilea, Destiny. Membership: Merck Co-Chair V710 Vaccine. Educational fees: Green Cross, Cubist, Cerexa, Durata, Theravance; Debiopharm. Royalties: UpToDate. MRY is a founder and shareholder of NovaDigm Therapeutics, Inc., which develops vaccines and immunotherapeutics targeting multi-drug-resistant pathogens, including S. aureus. He has received research funding from the U.S. National Institutes of Health and the U.S. Department of Defense and has received honoraria for educational activities or consultation from Alexion/AstraZeneca, Genentech-Roche and Horizon/Amgen. EFR is on the board of Federation of Clinical Immunology Societies. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be constructed as a potential conflict of interest., (Copyright © 2024 Parmar, Pickering, Ahn, Rossetti, Gjertson, Ruffin, Chan, Fowler, Yeaman, Reed and MRSA Systems Immunology Group.)
- Published
- 2024
- Full Text
- View/download PDF
18. A genome-scale screen identifies sulfated glycosaminoglycans as pivotal in epithelial cell damage by Candida albicans .
- Author
-
Lin J, Miao J, Schaefer KG, Russell CM, Pyron RJ, Zhang F, Phan QT, Solis-Swidergall NV, Liu H, Tashiro M, Dordick JS, Linhardt RJ, Yeaman MR, King GM, Barrera FN, Peters BM, and Filler SG
- Abstract
Candidalysin is a cytolytic peptide produced by the opportunistic fungal pathogen Candida albicans. This peptide is a key virulence factor in mouse models of mucosal and hematogenously disseminated candidiasis. Despite intense interest in the role of candidalysin in C. albicans pathogenicity, its host cell targets have remained elusive. To fill this knowledge gap, we performed a genome-wide loss-of-function CRISPR screen in a human oral epithelial cell line to identify specific host factors required for susceptibility to candidalysin-induced cellular damage. Among the top hits were XYLT2 , B3GALT6 and B3GAT3 , genes that function in glycosaminoglycan (GAG) biosynthesis. Deletion of these genes led to the absence of GAGs such as heparan sulfate on the epithelial cell surface and increased resistance to damage induced by both candidalysin and live C. albicans. Biophysical analyses including surface plasmon resonance and atomic force and electron microscopy indicated that candidalysin physically binds to sulfated GAGs, facilitating its oligomerization or enrichment on the host cell surface. The addition of exogenous sulfated GAGs or the GAG analogue dextran sulfate protected cells against candidalysin-induced damage. Dextran sulfate, but not non-sulfated dextran, also inhibited epithelial cell endocytosis of C. albicans and fungal-induced epithelial cell cytokine and chemokine production. In a murine model of vulvovaginal candidiasis, topical dextran sulfate administration reduced host tissue damage and decreased intravaginal IL-1β and neutrophil levels. Collectively, these data indicate that GAGs are epithelial cell targets of candidalysin and can be used therapeutically to protect cells from candidalysin-induced damage.
- Published
- 2024
- Full Text
- View/download PDF
19. Use of Transcriptional Signatures to Differentiate Pathogen-Specific and Treatment-Specific Host Responses in Patients With Bacterial Bloodstream Infections.
- Author
-
Thaden JT, Ahn R, Ruffin F, Gjertson DW, Hoffmann A, Fowler VG Jr, and Yeaman MR
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Transcriptome, Klebsiella pneumoniae genetics, Escherichia coli genetics, Adult, Staphylococcus aureus genetics, Staphylococcus aureus drug effects, Host-Pathogen Interactions genetics, Host-Pathogen Interactions immunology, Gene Expression Profiling, Methicillin-Resistant Staphylococcus aureus genetics, Methicillin-Resistant Staphylococcus aureus drug effects, Sequence Analysis, RNA, Bacteremia microbiology, Bacteremia drug therapy, Bacteremia immunology, Anti-Bacterial Agents therapeutic use, Anti-Bacterial Agents pharmacology
- Abstract
Background: Clinical outcomes in bacterial bloodstream infections (BSIs) are influenced by bacterial species, host immunity, and antibiotic therapy. The mechanisms by which such factors influence outcomes are poorly understood. We aimed to identify bacterial- and antibiotic-specific host transcriptional signatures in patients with bacterial BSI., Methods: RNA sequencing was performed on blood samples from patients with BSI due to gram-negative (GN) versus gram-positive (GP) pathogens: Escherichia coli (n = 30) or Klebsiella pneumoniae (n = 28) versus methicillin-susceptible Staphylococcus aureus (MSSA) (n = 24) or methicillin-resistant S. aureus (MRSA) (n = 58). Patients were matched by age, sex, and race., Results: No significant host transcriptome differences were detected in patients with E. coli versus K. pneumoniae BSI, so these were considered together as GN BSI. Relative to S. aureus BSI, patients with GN BSI had increased activation of the classic complement system. However, the most significant signal was a reduction in host transcriptional signatures involving mitochondrial energy transduction and oxidative burst in MRSA versus MSSA. This attenuated host transcriptional signature remained after controlling for antibiotic therapy., Conclusions: Given the importance of immune cellular energetics and reactive oxygen species in eliminating hematogenous or intracellular MRSA, these findings may offer insights into its persistence relative to other bacterial BSIs., Competing Interests: Potential conflicts of interest. J. T. T. reports being a scientific advisor for Resonantia Diagnostics. V. G. F. reports personal fees from Novartis, Debiopharm, Genentech, Achaogen, Affinium, The Medicines Company, MedImmune, Bayer, Basilea, Affinergy, Janssen, Contrafect, Regeneron, Destiny, Amphliphi Biosciences, Integrated Biotherapeutics, C3J, Armata, Valanbio, Akagera, Aridis, GSK, and Roche; grants from the National Institutes of Health, AstraZeneca/MedImmune, Allergan, Pfizer, Advanced Liquid Logics, Theravance, Novartis, Merck, Medical Biosurfaces, Locus, Affinergy, Contrafect, Karius, Genentech, Regeneron, Deep Blue, Basilea, and Janssen; royalties from UpToDate; stock options from Valanbio and ArcBio; honoraria from the Infectious Diseases Society of America for his service as associate editor of Clinical Infectious Diseases; and a sepsis diagnostics patent pending. M. R. Y. reports honoraria for academic educational services from Alexion/Astrazeneca, Horizon/Amgen, and Genentech-Roche. All other authors report no potential conflicts. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
20. Tensor modeling of MRSA bacteremia cytokine and transcriptional patterns reveals coordinated, outcome-associated immunological programs.
- Author
-
Chin JL, Tan ZC, Chan LC, Ruffin F, Parmar R, Ahn R, Taylor SD, Bayer AS, Hoffmann A, Fowler VG Jr, Reed EF, Yeaman MR, and Meyer AS
- Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) bacteremia is a common and life-threatening infection that imposes up to 30% mortality even when appropriate therapy is used. Despite in vitro efficacy determined by minimum inhibitory concentration breakpoints, antibiotics often fail to resolve these infections in vivo, resulting in persistent MRSA bacteremia. Recently, several genetic, epigenetic, and proteomic correlates of persistent outcomes have been identified. However, the extent to which single variables or their composite patterns operate as independent predictors of outcome or reflect shared underlying mechanisms of persistence is unknown. To explore this question, we employed a tensor-based integration of host transcriptional and cytokine datasets across a well-characterized cohort of patients with persistent or resolving MRSA bacteremia outcomes. This method yielded high correlative accuracy with outcomes and immunologic signatures united by transcriptomic and cytokine datasets. Results reveal that patients with persistent MRSA bacteremia (PB) exhibit signals of granulocyte dysfunction, suppressed antigen presentation, and deviated lymphocyte polarization. In contrast, patients with resolving bacteremia (RB) heterogeneously exhibit correlates of robust antigen-presenting cell trafficking and enhanced neutrophil maturation corresponding to appropriate T lymphocyte polarization and B lymphocyte response. These results suggest that transcriptional and cytokine correlates of PB vs. RB outcomes are complex and may not be disclosed by conventional modeling. In this respect, a tensor-based integration approach may help to reveal consensus molecular and cellular mechanisms and their biological interpretation., (© The Author(s) 2024. Published by Oxford University Press on behalf of National Academy of Sciences.)
- Published
- 2024
- Full Text
- View/download PDF
21. Anti-aquaporin-4 immune complex stimulates complement-dependent Th17 cytokine release in neuromyelitis optica spectrum disorders.
- Author
-
Nishiyama S, Seok JM, Wright AE, Lotan I, Mikami T, Drosu NC, Bobrowski-Khoury N, Anderson MR, Bilodeau PA, Schindler P, Paul F, Aoki M, Yeaman MR, and Levy M
- Subjects
- Humans, Cytokines metabolism, Antigen-Antibody Complex metabolism, Leukocytes, Mononuclear metabolism, Interleukin-17 metabolism, Interleukin-6 metabolism, Rituximab pharmacology, Rituximab therapeutic use, Rituximab metabolism, Autoantibodies, Aquaporin 4, Complement System Proteins metabolism, Immunoglobulin G metabolism, Neuromyelitis Optica
- Abstract
Proinflammatory cytokines, such as (IL: interleukin) IL-6 and IL-17A, and complement fixation are critical in the immunopathogenesis of neuromyelitis optica spectrum disorders (NMOSD). Blocking the IL-6 receptor or the C5 complement pathway reduces relapse risk. However, the role of interleukin (IL)-6 and complement in aquaporin-4 (AQP4) autoimmunity remains unclear. To investigate the role of the anti-AQP4 immunoglobulin (AQP4-IgG)/AQP4 immunocomplex on the induction and profile of ex vivo cytokine and surface marker expression in peripheral blood mononuclear cells (PBMC) culture. Isolated PBMCs obtained from 18 patients with AQP4-IgG-seropositive-NMOSD (8 treatment-naive, 10 rituximab-treated) or ten healthy controls were cultured with AQP4-immunocomplex with or without complement. Changes in PBMC surface markers and cytokine expression were profiled using flow cytometry and ELISA. PBMCs derived from treatment-naive NMOSD patients stimulated with a complex mixture of serum complement proteins produced significant elevations of IL-17A and IL-6. Rituximab-treated patients also exhibited higher IL-6 but not IL-17A release. IL-6 and IL-17A elevations are not observed without complement. Co-stimulation of PBMCs with AQP4-IgG/AQP4 immunocomplex and complement prompts a Th17-biased response consistent with the inflammatory paradigm observed in NMOSD. A possible inflammation model is proposed via antigen-specific autoreactive peripheral blood cells, including NK/NKT cells., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
22. Viral afterlife: SARS-CoV-2 as a reservoir of immunomimetic peptides that reassemble into proinflammatory supramolecular complexes.
- Author
-
Zhang Y, Bharathi V, Dokoshi T, de Anda J, Ursery LT, Kulkarni NN, Nakamura Y, Chen J, Luo EWC, Wang L, Xu H, Coady A, Zurich R, Lee MW, Matsui T, Lee H, Chan LC, Schepmoes AA, Lipton MS, Zhao R, Adkins JN, Clair GC, Thurlow LR, Schisler JC, Wolfgang MC, Hagan RS, Yeaman MR, Weiss TM, Chen X, Li MMH, Nizet V, Antoniak S, Mackman N, Gallo RL, and Wong GCL
- Subjects
- Humans, Animals, Mice, Endothelial Cells, Proteome, Peptides, SARS-CoV-2, COVID-19
- Abstract
It is unclear how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leads to the strong but ineffective inflammatory response that characterizes severe Coronavirus disease 2019 (COVID-19), with amplified immune activation in diverse cell types, including cells without angiotensin-converting enzyme 2 receptors necessary for infection. Proteolytic degradation of SARS-CoV-2 virions is a milestone in host viral clearance, but the impact of remnant viral peptide fragments from high viral loads is not known. Here, we examine the inflammatory capacity of fragmented viral components from the perspective of supramolecular self-organization in the infected host environment. Interestingly, a machine learning analysis to SARS-CoV-2 proteome reveals sequence motifs that mimic host antimicrobial peptides (xenoAMPs), especially highly cationic human cathelicidin LL-37 capable of augmenting inflammation. Such xenoAMPs are strongly enriched in SARS-CoV-2 relative to low-pathogenicity coronaviruses. Moreover, xenoAMPs from SARS-CoV-2 but not low-pathogenicity homologs assemble double-stranded RNA (dsRNA) into nanocrystalline complexes with lattice constants commensurate with the steric size of Toll-like receptor (TLR)-3 and therefore capable of multivalent binding. Such complexes amplify cytokine secretion in diverse uninfected cell types in culture (epithelial cells, endothelial cells, keratinocytes, monocytes, and macrophages), similar to cathelicidin's role in rheumatoid arthritis and lupus. The induced transcriptome matches well with the global gene expression pattern in COVID-19, despite using <0.3% of the viral proteome. Delivery of these complexes to uninfected mice boosts plasma interleukin-6 and CXCL1 levels as observed in COVID-19 patients., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
23. Prevalence, Demographic, and Clinical Factors Associated With Cognitive Dysfunction in Patients With Neuromyelitis Optica Spectrum Disorder.
- Author
-
Vlahovic L, McDonald J, Hinman J, Tomczak A, Lock C, Palmer CA, Cook LJ, Yeaman MR, Burnett MK, Deutsch GK, Nelson LM, and Han MH
- Subjects
- Humans, Prevalence, Retrospective Studies, Rituximab, Neoplasm Recurrence, Local, Aquaporin 4, Neuromyelitis Optica complications, Neuromyelitis Optica epidemiology, Cognitive Dysfunction epidemiology
- Abstract
Background and Objectives: Neuromyelitis optica spectrum disorder (NMOSD) is a chronic CNS demyelinating autoimmune disorder targeting the astrocyte antigen aquaporin-4 (AQP4), typically presenting with optic neuritis, transverse myelitis, and brain syndromes. Cognitive dysfunction (CD) in NMOSD is under-recognized and poorly understood. The purpose of this study was to evaluate the prevalence and clinical variables associated with CD in NMOSD., Methods: This observational retrospective study with longitudinal follow-up describes a clinical cohort seen in the Collaborative International Research in Clinical and Longitudinal Experience Study in NMOSD. Serial Montreal Cognitive Assessments (MoCAs) were performed upon enrollment and at 6-month intervals to evaluate longitudinal cognitive function relative to demographic and disease-related factors. We used 2-tailed t test, analysis of variance, the χ
2 test, linear regression for univariable and adjusted analyses and simultaneous linear regression and mixed-effects model for multivariable analyses., Results: Thirty-four percent (75/219) of patients met criteria for CD (MoCA <26); 29% (64/219) showed mild dysfunction (MoCA 20-26/30), and 5% (11/219) showed moderate (MoCA <20/30) dysfunction. Patients with less neurologic disability and lower pain scores had higher MoCA scores (95% CI 0.24-0.65 and 95% CI 0.09-0.42, respectively). Patients with at least high school education scored higher on the MoCA (95% CI 2.2-5). When comparing patients dichotomized for CD, patients never on rituximab scored higher than patients only treated with rituximab ( p < 0.029). There was no significant association between annualized relapse rate, age, sex, disease duration, AQP4 serostatus or brain lesions, and CD. CD was more pronounced among Black than White patients (95% CI -2.7 to -0.7). Multivariable analysis of serial MoCA did not indicate change ( p = 0.715). Descriptive analysis of serial MoCA showed 30% (45/150) of patients with worsening MoCA performance had impaired language and verbal recall., Discussion: To our knowledge, this is the largest study of diverse cohort to investigate CD in patients with NMOSD. Our findings demonstrate 34% of patients with NMOSD experience mild-to-moderate CD, while 30% of patients demonstrated decline on serial testing. The substantial prevalence of CD in this pilot report highlights the need for improved and validated screening tools and comprehensive measures to investigate CD in NMOSD.- Published
- 2024
- Full Text
- View/download PDF
24. Retinal Changes After Acute and Late Optic Neuritis in Aquaporin-4 Antibody Seropositive NMOSD.
- Author
-
Oertel FC, Zimmermann HG, Motamedi S, Bereuter C, Asseyer ES, Chien C, Marignier R, Cobo-Calvo A, Leocani L, Pisa M, Radaelli M, Villoslada P, Sanchez-Dalmau B, Martinez-Lapiscina EH, Lana-Peixoto MA, Fontenelle MA, Aktas O, Ringelstein M, Albrecht P, Green AJ, Yeaman MR, Smith TJ, Cook L, Paul F, and Brandt AU
- Abstract
Competing Interests: F. C. Oertel was an employee of Nocturne GmbH and receives research support by the American Academy of Neurology and National Multiple Sclerosis Society (US), unrelated to this work and funding by the German Association of Neurology (Deutsche Gesellschaft für Neurologie) in context of this project. H. G. Zimmermann reports speaking honoraria and grants from Novartis and speaking honoraria from Bayer Healthcare, unrelated to this study. C. Chien has received speaking honoraria from Bayer and research funding from Novartis, unrelated to this study. R. Marignier serves on the scientific advisory board for MedImmune and has received funding for travel and honoraria from Biogen, Merck Serono, Novartis, Sanofi- Genzyme, Roche and Teva. A. Cobo-Calvo received funding from the Instituto de Salud Carlos III (Spain) JR19/00007 unrelated to this manuscript. L. Leocani received honoraria for consulting services from Merck, Roche, Biogen and for speaking activities from Teva; research support from Merck, Biogen, Novartis; travel support from Merck, Roche, Biogen, Almirall. M. Radaelli received speaker honoraria from Novartis, Bayer, Roche, Alexion and Ipsen and travel reimbursement from Bayer, Biogen, Merz, Genzyme, Teva, Roche and Merck, none related to this study. P. Villoslada hold stocks and has received consultancy fees from Accure Therapeutics, QMENTA, CLight, NeuroPrex, Attune Neurosciences, Spiral Therapeutics and Adhera Health. M. A. Lana-Peixoto has received funding for travel and speaker honoraria from Novartis, Sanofi- Genzyme, and Roche. O. Aktas has received honoraria for speaking/consultation and travel grants from Bayer Healthcare, Biogen Idec, Chugai, Novartis, Medimmune, Merck Serono, and Teva and research grants from Bayer Healthcare, Biogen Idec, Novartis, and Teva, not related to this study. M. Ringelstein received speaker honoraria from Novartis, Bayer, Roche, Alexion, and Ipsen, and travel reimbursement from Bayer, Biogen, Merz, Genzyme, Teva, Roche, and Merck, none related to this study. P. Albrecht reports grants, personal fees, and nonfinancial support from Allergan, Biogen, Ipsen, Merz Pharmaceuticals, Novartis, and Roche, personal fees and nonfinancial support from Bayer Healthcare, and Merck, and nonfinancial support from Sanofi-Aventis/Genzyme. M. R. Yeaman is the founder and a shareholder of NovaDigm Therapeutics, Inc.; he receives funding from the United States National Institutes of Health and United States Department of Defense; he holds U.S. and international patents on immunotherapeutic and anti-infective technologies, is a member of the Genentech-Roche Scientific Advisory Committee and adviser to The Guthy-Jackson Charitable Foundation. T. J. Smith was issued US patents covering the therapeutic targeting of IGF-I receptor in autoimmune diseases. He is a paid consultant for Horizon Thera and Immunovant and is a scientific advisor to the Guthy-Jackson Charitable Foundation. He receives research funding from the National Institutes of Health. F. Paul reports research grants and speaker honoraria from Bayer, Teva, Genzyme, Merck, Novartis, MedImmune and is member of the steering committee of the OCTIMS study (Novartis), all unrelated to this work. A. U. Brandt is cofounder and shareholder of Motognosis and Nocturne. He is named as inventor on several patent applications regarding MS serum biomarkers, OCT image analysis and perceptive visual computing. E. H. Martinez-Lapiscina, S. Motamedi, M. Pisa, SvS, C. Bereuter, and M. A. Fontenelle report no conflicts of interest.
- Published
- 2023
- Full Text
- View/download PDF
25. Diflunisal and Analogue Pharmacophores Mediating Suppression of Virulence Phenotypes in Staphylococcus aureus .
- Author
-
Chan LC, Lee HK, Wang L, Chaili S, Xiong YQ, Bayer AS, Proctor RA, and Yeaman MR
- Abstract
Invasive methicillin-resistant Staphylococcus aureus (MRSA) infections are leading causes of morbidity and mortality that are complicated by increasing resistance to conventional antibiotics. Thus, minimizing virulence and enhancing antibiotic efficacy against MRSA is a public health imperative. We originally demonstrated that diflunisal (DIF; [2-hydroxy-5-(2,4-difluorophenyl) benzoic acid]) inhibits S. aureus virulence factor expression. To investigate pharmacophores that are active in this function, we evaluated a library of structural analogues for their efficacy to modulate virulence phenotypes in a panel of clinically relevant S. aureus isolates in vitro. Overall, the positions of the phenyl, hydroxyl, and carboxylic moieties and the presence or type of halogen (F vs. Cl) influenced the efficacy of compounds in suppressing hemolysis, proteolysis, and biofilm virulence phenotypes. Analogues lacking halogens inhibited proteolysis to an extent similar to DIF but were ineffective at reducing hemolysis or biofilm production. In contrast, most analogues lacking the hydroxyl or carboxylic acid groups did not suppress proteolysis but did mitigate hemolysis and biofilm production to an extent similar to DIF. Interestingly, chirality and the substitution of fluorine with chlorine resulted in a differential reduction in virulence phenotypes. Together, this pattern of data suggests virulence-suppressing pharmacophores of DIF and structural analogues integrate halogen, hydroxyl, and carboxylic acid moiety stereochemistry. The anti-virulence effects of DIF were achieved using concentrations that are safe in humans, do not impair platelet antimicrobial functions, do not affect S. aureus growth, and do not alter the efficacy of conventional antibiotics. These results offer proof of concept for using novel anti-virulence strategies as adjuvants to antibiotic therapy to address the challenge of MRSA infection.
- Published
- 2023
- Full Text
- View/download PDF
26. Diagnostic value of intereye difference metrics for optic neuritis in aquaporin-4 antibody seropositive neuromyelitis optica spectrum disorders.
- Author
-
Oertel FC, Zimmermann HG, Motamedi S, Chien C, Aktas O, Albrecht P, Ringelstein M, Dcunha A, Pandit L, Martinez-Lapiscina EH, Sanchez-Dalmau B, Villoslada P, Palace J, Roca-Fernández A, Leite MI, Sharma SM, Leocani L, Pisa M, Radaelli M, Lana-Peixoto MA, Fontenelle MA, Havla J, Ashtari F, Kafieh R, Dehghani A, Pourazizi M, Marignier R, Cobo-Calvo A, Asgari N, Jacob A, Huda S, Mao-Draayer Y, Green AJ, Kenney R, Yeaman MR, Smith TJ, Cook L, Brandt AU, Paul F, and Petzold A
- Subjects
- Humans, Retrospective Studies, Benchmarking, Tomography, Optical Coherence methods, Autoantibodies, Aquaporin 4, Neuromyelitis Optica diagnosis, Optic Neuritis diagnosis, Aquaporins
- Abstract
Background: The novel optic neuritis (ON) diagnostic criteria include intereye differences (IED) of optical coherence tomography (OCT) parameters. IED has proven valuable for ON diagnosis in multiple sclerosis but has not been evaluated in aquaporin-4 antibody seropositive neuromyelitis optica spectrum disorders (AQP4+NMOSD). We evaluated the diagnostic accuracy of intereye absolute (IEAD) and percentage difference (IEPD) in AQP4+NMOSD after unilateral ON >6 months before OCT as compared with healthy controls (HC)., Methods: Twenty-eight AQP4+NMOSD after unilateral ON (NMOSD-ON), 62 HC and 45 AQP4+NMOSD without ON history (NMOSD-NON) were recruited by 13 centres as part of the international Collaborative Retrospective Study on retinal OCT in Neuromyelitis Optica study. Mean thickness of peripapillary retinal nerve fibre layer (pRNFL) and macular ganglion cell and inner plexiform layer (GCIPL) were quantified by Spectralis spectral domain OCT. Threshold values of the ON diagnostic criteria (pRNFL: IEAD 5 µm, IEPD 5%; GCIPL: IEAD: 4 µm, IEPD: 4%) were evaluated using receiver operating characteristics and area under the curve (AUC) metrics., Results: The discriminative power was high for NMOSD-ON versus HC for IEAD (pRNFL: AUC 0.95, specificity 82%, sensitivity 86%; GCIPL: AUC 0.93, specificity 98%, sensitivity 75%) and IEPD (pRNFL: AUC 0.96, specificity 87%, sensitivity 89%; GCIPL: AUC 0.94, specificity 96%, sensitivity 82%). The discriminative power was high/moderate for NMOSD-ON versus NMOSD-NON for IEAD (pRNFL: AUC 0.92, specificity 77%, sensitivity 86%; GCIP: AUC 0.87, specificity 85%, sensitivity 75%) and for IEPD (pRNFL: AUC 0.94, specificity 82%, sensitivity 89%; GCIP: AUC 0.88, specificity 82%, sensitivity 82%)., Conclusions: Results support the validation of the IED metrics as OCT parameters of the novel diagnostic ON criteria in AQP4+NMOSD., Competing Interests: Competing interests: The authors have stated explicitly that there are no conflicts of interest in connection with this article., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
27. Diflunisal Attenuates Virulence Factor Gene Regulation and Phenotypes in Staphylococcus aureus .
- Author
-
Chan LC, Park M, Lee HK, Chaili S, Xiong YQ, Bayer AS, Proctor RA, and Yeaman MR
- Abstract
Virulence factor expression is integral to pathogenicity of Staphylococcus aureus . We previously demonstrated that aspirin, through its major metabolite, salicylic acid (SAL), modulates S. aureus virulence phenotypes in vitro and in vivo. We compared salicylate metabolites and a structural analogue for their ability to modulate S. aureus virulence factor expression and phenotypes: (i) acetylsalicylic acid (ASA, aspirin); (ii) ASA metabolites, salicylic acid (SAL), gentisic acid (GTA) and salicyluric acid (SUA); or (iii) diflunisal (DIF), a SAL structural analogue. None of these compounds altered the growth rate of any strain tested. ASA and its metabolites SAL, GTA and SUA moderately impaired hemolysis and proteolysis phenotypes in multiple S. aureus strain backgrounds and their respective deletion mutants. Only DIF significantly inhibited these virulence phenotypes in all strains. The kinetic profiles of ASA, SAL or DIF on expression of hla (alpha hemolysin), sspA (V8 protease) and their regulators ( sigB , sarA , agr (RNAIII)) were assessed in two prototypic strain backgrounds: SH1000 (methicillin-sensitive S. aureus ; MSSA) and LAC-USA300 (methicillin-resistant S. aureus ; MRSA). DIF induced sigB expression which is coincident with the significant inhibition of RNAIII expression in both strains and precedes significant reductions in hla and sspA expression. The inhibited expression of these genes within 2 h resulted in the durable suppression of hemolysis and proteolysis phenotypes. These results indicate that DIF modulates the expression of key virulence factors in S. aureus via a coordinated impact on their relevant regulons and target effector genes. This strategy may hold opportunities to develop novel antivirulence strategies to address the ongoing challenge of antibiotic-resistant S. aureus .
- Published
- 2023
- Full Text
- View/download PDF
28. Tensor-based insights into systems immunity and infectious disease.
- Author
-
Chin JL, Chan LC, Yeaman MR, and Meyer AS
- Subjects
- Humans, Communicable Diseases, Immunity
- Abstract
Profiling immune responses across several dimensions, including time, patients, molecular features, and tissue sites, can deepen our understanding of immunity as an integrated system. These studies require new analytical approaches to realize their full potential. We highlight recent applications of tensor methods and discuss several future opportunities., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
29. Longitudinal analysis of SARS-CoV-2 infection and vaccination in the LA-SPARTA cohort reveals increased risk of infection in vaccinated Hispanic participants.
- Author
-
Jenkins MM, Phan Tran D, Flores EA, Kupferwasser D, Pickering H, Zheng Y, Gjertson DW, Ross TM, Schaenman JM, Miller LG, Yeaman MR, and Reed EF
- Subjects
- Humans, Antibodies, Ethnicity, Hispanic or Latino, Nucleocapsid Proteins, SARS-CoV-2, COVID-19 Vaccines, Occupational Exposure, COVID-19 epidemiology, COVID-19 prevention & control, Vaccination
- Abstract
Introduction: SARS-CoV-2 is the etiologic agent of coronavirus disease 2019 (COVID-19). Questions remain regarding correlates of risk and immune protection against COVID-19., Methods: We prospectively enrolled 200 participants with a high risk of SARS-CoV-2 occupational exposure at a U.S. medical center between December 2020 and April 2022. Participant exposure risks, vaccination/infection status, and symptoms were followed longitudinally at 3, 6, and 12 months, with blood and saliva collection. Serological response to the SARS-CoV-2 spike holoprotein (S), receptor binding domain (RBD) and nucleocapsid proteins (NP) were quantified by ELISA assay., Results: Based on serology, 40 of 200 (20%) participants were infected. Healthcare and non-healthcare occupations had equivalent infection incidence. Only 79.5% of infected participants seroconverted for NP following infection, and 11.5% were unaware they had been infected. The antibody response to S was greater than to RBD. Hispanic ethnicity was associated with 2-fold greater incidence of infection despite vaccination in this cohort., Discussion: Overall, our findings demonstrate: 1) variability in the antibody response to SARS-CoV-2 infection despite similar exposure risk; 2) the concentration of binding antibody to the SARS-CoV-2 S or RBD proteins is not directly correlated with protection against infection in vaccinated individuals; and 3) determinants of infection risk include Hispanic ethnicity despite vaccination and similar occupational exposure., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Jenkins, Phan Tran, Flores, Kupferwasser, Pickering, Zheng, Gjertson, Ross, Schaenman, Miller, Yeaman and Reed.)
- Published
- 2023
- Full Text
- View/download PDF
30. Clinical and epidemiological correlates of treatment change in patients with NMOSD: insights from the CIRCLES cohort.
- Author
-
Gholizadeh S, Exuzides A, Lewis KE, Palmer C, Waltz M, Rose JW, Jolley AM, Behne JM, Behne MK, Blaschke TF, Smith TJ, Sinnott J, Cook LJ, and Yeaman MR
- Subjects
- Humans, Quality of Life, Neoplasm Recurrence, Local, Spinal Cord, Longitudinal Studies, Aquaporin 4, Retrospective Studies, Autoantibodies, Neuromyelitis Optica drug therapy, Neuromyelitis Optica epidemiology
- Abstract
Objective: Neuromyelitis optica spectrum disorders (NMOSD) represent rare autoimmune diseases of the central nervous system largely targeting optic nerve(s) and spinal cord. The present analysis used real-world data to identify clinical and epidemiological correlates of treatment change in patients with NMOSD., Methods: CIRCLES is a longitudinal, observational study of NMOSD conducted at 15 centers across North America. Patients with ≥ 60 days of follow-up and receiving on-study maintenance treatment were evaluated. The mean annual relapse rate (ARR) was estimated using negative binomial models; the likelihood of treatment change was estimated using Cox proportional hazards models. Relapses were included as time-varying covariates to estimate the relationship to treatment change., Results: Of 542 patients included, 171 (31.5%) experienced ≥ 1 relapse on the study and 133 patients (24.5%) had ≥ 1 change in the treatment regimen. Two categories of variables significantly correlated with the likelihood of treatment change: (1) relapse: any on-study relapse (hazard ratio [HR] = 2.91; p < 0.001), relapse phenotypes (HR range = 2.15-5.49; p < 0.001), and pre-study ARR > 0.75 (HR 2.28; p < 0.001); 2) disease phenotype: brain syndrome only vs transverse myelitis involvement at onset (HR 2.44; p = 0.008), disease duration < 1 vs > 5 years (HR 1.66; p = 0.028), or autoimmune comorbidity (HR 1.55; p = 0.015). A subset of these factors significantly correlated with shorter time to first rituximab discontinuation., Conclusions: In CIRCLES, relapse patterns and disease phenotype significantly correlated with changes in the maintenance treatment regimen. Such findings may facilitate the identification of patients with NMOSD who are likely to benefit from treatment change to reduce relapse risk or disease burden and enhance the quality of life., (© 2022. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
31. Developing a standardized but extendable framework to increase the findability of infectious disease datasets.
- Author
-
Tsueng G, Cano MAA, Bento J, Czech C, Kang M, Pache L, Rasmussen LV, Savidge TC, Starren J, Wu Q, Xin J, Yeaman MR, Zhou X, Su AI, Wu C, Brown L, Shabman RS, and Hughes LD
- Subjects
- Metadata, Reproducibility of Results, Humans, Communicable Diseases, Datasets as Topic standards
- Abstract
Biomedical datasets are increasing in size, stored in many repositories, and face challenges in FAIRness (findability, accessibility, interoperability, reusability). As a Consortium of infectious disease researchers from 15 Centers, we aim to adopt open science practices to promote transparency, encourage reproducibility, and accelerate research advances through data reuse. To improve FAIRness of our datasets and computational tools, we evaluated metadata standards across established biomedical data repositories. The vast majority do not adhere to a single standard, such as Schema.org, which is widely-adopted by generalist repositories. Consequently, datasets in these repositories are not findable in aggregation projects like Google Dataset Search. We alleviated this gap by creating a reusable metadata schema based on Schema.org and catalogued nearly 400 datasets and computational tools we collected. The approach is easily reusable to create schemas interoperable with community standards, but customized to a particular context. Our approach enabled data discovery, increased the reusability of datasets from a large research consortium, and accelerated research. Lastly, we discuss ongoing challenges with FAIRness beyond discoverability., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
32. MRSA Isolates from Patients with Persistent Bacteremia Generate Nonstable Small Colony Variants In Vitro within Macrophages and Endothelial Cells during Prolonged Vancomycin Exposure.
- Author
-
Fauerharmel-Nunes T, Flannagan RS, Goncheva MI, Bayer AS, Fowler VG Jr, Chan LC, Yeaman MR, Xiong YQ, and Heinrichs DE
- Subjects
- Humans, Vancomycin pharmacology, Methicillin Resistance, Endothelial Cells, Microbial Sensitivity Tests, Macrophages, Anti-Bacterial Agents pharmacology, Anti-Bacterial Agents therapeutic use, Methicillin-Resistant Staphylococcus aureus, Staphylococcal Infections drug therapy, Bacteremia drug therapy
- Abstract
Staphylococcus aureus (especially methicillin-resistant S. aureus [MRSA]) is frequently associated with persistent bacteremia (PB) during vancomycin therapy despite consistent susceptibility in vitro . Strategic comparisons of PB strains versus those from vancomycin-resolving bacteremia (RB) would yield important mechanistic insights into PB outcomes. Clinical PB versus RB isolates were assessed in vitro for intracellular replication and small colony variant (SCV) formation within macrophages and endothelial cells (ECs) in the presence or absence of exogenous vancomycin. In both macrophages and ECs, PB and RB isolates replicated within lysosome-associated membrane protein-1 (LAMP-1)-positive compartments. PB isolates formed nonstable small colony variants (nsSCVs) in vancomycin-exposed host cells at a significantly higher frequency than matched RB isolates (in granulocyte-macrophage colony-stimulating factor [GM-CSF], human macrophages PB versus RB, P < 0.0001 at 48 h; in ECs, PB versus RB, P < 0.0001 at 24 h). This phenotype could represent one potential basis for the unique ability of PB isolates to adaptively resist vancomycin therapy and cause PB in humans. Elucidating the molecular mechanism(s) by which PB strains form nsSCVs could facilitate the discovery of novel treatment strategies to mitigate PB due to MRSA.
- Published
- 2023
- Full Text
- View/download PDF
33. Staphylococcus aureus adaptive evolution: Recent insights on how immune evasion, immunometabolic subversion and host genetics impact vaccine development.
- Author
-
Wong Fok Lung T, Chan LC, Prince A, Yeaman MR, Archer NK, Aman MJ, and Proctor RA
- Subjects
- Humans, Staphylococcus aureus genetics, Immune Evasion, Prospective Studies, Staphylococcal Infections microbiology, Vaccines
- Abstract
Despite meritorious attempts, a S. aureus vaccine that prevents infection or mitigates severity has not yet achieved efficacy endpoints in prospective, randomized clinical trials. This experience underscores the complexity of host- S. aureus interactions, which appear to be greater than many other bacterial pathogens against which successful vaccines have been developed. It is increasingly evident that S. aureus employs strategic countermeasures to evade or exploit human immune responses. From entering host cells to persist in stealthy intracellular reservoirs, to sensing the environmental milieu and leveraging bacterial or host metabolic products to reprogram host immune responses, S. aureus poses considerable challenges for the development of effective vaccines. The fact that this pathogen causes distinct types of infections and can undergo transient genetic, transcriptional or metabolic adaptations in vivo that do not occur in vitro compounds challenges in vaccine development. Notably, the metabolic versatility of both bacterial and host immune cells as they compete for available substrates within specific tissues inevitably impacts the variable repertoire of gene products that may or may not be vaccine antigens. In this respect, S. aureus has chameleon phenotypes that have alluded vaccine strategies thus far. Nonetheless, a number of recent studies have also revealed important new insights into pathogenesis vulnerabilities of S. aureus . A more detailed understanding of host protective immune defenses versus S. aureus adaptive immune evasion mechanisms may offer breakthroughs in the development of effective vaccines, but at present this goal remains a very high bar. Coupled with the recent advances in human genetics and epigenetics, newer vaccine technologies may enable such a goal. If so, future vaccines that protect against or mitigate the severity of S. aureus infections are likely to emerge at the intersection of precision and personalized medicine. For now, the development of S. aureus vaccines or alternative therapies that reduce mortality and morbidity must continue to be pursued., Competing Interests: RP is a consultant for IBT which is producing a multi-valent antitoxin vaccine and is a member of the review board for the University of Rochester which is involved in an anti-Gmd vaccine effort. MY is a consultant for Genentech-Roche, Alexion/AstraZeneca and Horizon Pharmaceuticals, which are involved in discovery and development of immunotherapeutic agents and strategies. NA is a consultant for Janssen Pharmaceuticals and has received previous grant support from Pfizer and Boehringer Ingelheim, which are involved in discovery and development of immunotherapeutic agents and strategies. MA has stocks in Integrated Biotherapeutics, a company engaged in development of vaccines and antibody therapies for bacterial infections. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Wong Fok Lung, Chan, Prince, Yeaman, Archer, Aman and Proctor.)
- Published
- 2022
- Full Text
- View/download PDF
34. Long-term safety of satralizumab in neuromyelitis optica spectrum disorder (NMOSD) from SAkuraSky and SAkuraStar.
- Author
-
Yamamura T, Weinshenker B, Yeaman MR, De Seze J, Patti F, Lobo P, von Büdingen HC, Kou X, Weber K, and Greenberg B
- Subjects
- Humans, Double-Blind Method, Neuromyelitis Optica drug therapy, Antibodies, Monoclonal, Humanized adverse effects
- Abstract
Background: This analysis evaluated long-term safety findings from the SAkuraSky and SAkuraStar studies with satralizumab in patients with neuromyelitis optica spectrum disorder (NMOSD)., Methods: SAkuraSky (satralizumab in combination with baseline immunosuppressive therapy; IST) and SAkuraStar (satralizumab monotherapy) are international, multicenter, randomized, placebo-controlled, phase 3 studies consisting of a double-blind (DB) period followed by an open-label extension (OLE). The overall satralizumab treatment (OST) period safety population comprised patients receiving ≥1 dose of satralizumab in the DB and/or OLE periods (cut-off date: 22 February 2021). Safety was evaluated in the DB and OST periods., Results: In the SAkuraSky DB period, patients received satralizumab (n = 41) or placebo (n = 42) in addition to stable baseline IST; 75 patients were included in the OST population. In the SAkuraStar DB period, 63 patients received satralizumab monotherapy and 32 received placebo; 91 patients were included in the OST population. Median treatment exposure in the OST period was 4.4 years (range 0.1-7.0) in SAkuraSky and 4.0 years (range 0.1-6.1) in SAkuraStar. Rates of adverse events (AEs per 100 patient-years) and serious AEs in the OST period were comparable with satralizumab and placebo in the DB periods of both studies. Similarly, overall rates of infections and serious infections were consistent between the OST and DB periods with satralizumab, with no increase in rates of infections or serious infections over time. In the OST periods, longer exposure to satralizumab was not associated with a higher risk of severe (grade ≥3) laboratory changes versus the DB periods. No deaths or anaphylactic reactions to treatment with satralizumab were reported during the OST periods of both studies., Conclusion: The safety profile of satralizumab as a monotherapy or in combination with IST was maintained in the OLE, and no new safety concerns versus the DB period were observed., Clinical Trial Registration: ClinicalTrials.gov identifiers: NCT02028884 (SAkuraSky) and NCT02073279 (SAkuraStar)., Competing Interests: Declaration of Competing Interest This study was sponsored by F. Hoffmann-La Roche. Takashi Yamamura served on scientific advisory boards for Biogen, Takeda, Sumitomo, Novartis, and Chugai. He receives consulting fees from Biogen, Takeda, Mitsubishi Tanabe, Novartis, Roche, and Chugai. He carried out contracted research in Mitsubishi Tanabe, Novartis, Chugai, Sanofi, Chiome Bioscience, and Miraca Holdings (within the contract period but no deposit). He received speaker honoraria from Chugai, Takeda, Biogen, and Sumitomo. Brian G. Weinshenker reports consulting fees from UCB Biosciences, Mitsubishi Tanabe, Genentech, and Roche, and speaking fees from Genentech, Roche, and Novartis; he participated on the Attack Adjudication Committee for Alexion and Horizon Therapeutics (formerly MedImmune/Viela Bio). He reports personal fees from Chugai. He has a patent NMO-IgG for diagnosis of neuromyelitis optica with royalties received from RSR Ltd, Oxford University, Hospices Civils de Lyon, and MVZ Labor PD Dr. Volkmann und Kollegen GbR. Michael R. Yeaman received grants from the US National Institutes of Health and the US Department of Defense, and consulting fees from Roche, Horizon, and Alexion. He is a founder and shareholder in NovaDigm Therapeutics, Inc. and Metacin, Inc. He serves on the Genentech-Roche Strategic Scientific Committee for NMOSD and is Chair Medical Advisor to the Guthy-Jackson Charitable Foundation for NMOSD. Jerome De Seze received grants and personal fees from Roche, personal fees from Chugai, and has served on advisory boards in the expert committee for the clinical trial conducted by Chugai. Francesco Patti has served on the scientific advisory boards for Almirall, Bayer, Biogen, Calgene, Merck, Novartis, Roche, Sanofi, and TEVA; he also received speaker honoraria from the aforementioned companies and research grants for his department from Biogen and Merck. Patricia Lobo is an employee of ApotheCom, who is paid to provide medical writing assistance for F. Hoffmann-La Roche Ltd. H.-Christian von Büdingen, Xiujing Kou, and Kristina Weber are employees of F. Hoffmann-La Roche Ltd. Benjamin Greenberg received consulting fees from Alexion, Novartis, EMD Serono, Viela Bio, Genentech/Roche, Greenwich Biosciences, Axon Advisors, Rubin Anders, ABCAM, Signant, IQVIA, Sandoz, Druggability Technologies, Genzyme, and Immunovant. He receives contracted research fees from Clene Nanomedicine. He receives royalties from UpToDate., (Copyright © 2022. Published by Elsevier B.V.)
- Published
- 2022
- Full Text
- View/download PDF
35. Longitudinal Retinal Changes in MOGAD.
- Author
-
Oertel FC, Sotirchos ES, Zimmermann HG, Motamedi S, Specovius S, Asseyer ES, Chien C, Cook L, Vasileiou E, Filippatou A, Calabresi PA, Saidha S, Pandit L, D'Cunha A, Outteryck O, Zéphir H, Pittock S, Flanagan EP, Bhatti MT, Rommer PS, Bsteh G, Zrzavy T, Kuempfel T, Aktas O, Ringelstein M, Albrecht P, Ayzenberg I, Pakeerathan T, Knier B, Aly L, Asgari N, Soelberg K, Marignier R, Tilikete CF, Cobo Calvo A, Villoslada P, Sanchez-Dalmau B, Martinez-Lapiscina EH, Llufriu S, Green AJ, Yeaman MR, Smith TJ, Brandt AU, Chen J, Paul F, and Havla J
- Subjects
- Case-Control Studies, Cohort Studies, Humans, Longitudinal Studies, Optic Neuritis diagnostic imaging, Optic Neuritis etiology, Retina diagnostic imaging, Retinal Neurons, Tomography, Optical Coherence methods, Immunologic Deficiency Syndromes complications, Myelin-Oligodendrocyte Glycoprotein immunology, Optic Neuritis complications, Retinal Degeneration etiology
- Abstract
Objective: Patients with myelin oligodendrocyte glycoprotein antibody (MOG-IgG)-associated disease (MOGAD) suffer from severe optic neuritis (ON) leading to retinal neuro-axonal loss, which can be quantified by optical coherence tomography (OCT). We assessed whether ON-independent retinal atrophy can be detected in MOGAD., Methods: Eighty patients with MOGAD and 139 healthy controls (HCs) were included. OCT data was acquired with (1) Spectralis spectral domain OCT (MOGAD: N = 66 and HCs: N = 103) and (2) Cirrus high-definition OCT (MOGAD: N = 14 and HCs: N = 36). Macular combined ganglion cell and inner plexiform layer (GCIPL) and peripapillary retinal nerve fiber layer (pRNFL) were quantified., Results: At baseline, GCIPL and pRNFL were lower in MOGAD eyes with a history of ON (MOGAD-ON) compared with MOGAD eyes without a history of ON (MOGAD-NON) and HCs (p < 0.001). MOGAD-NON eyes had lower GCIPL volume compared to HCs (p < 0.001) in the Spectralis, but not in the Cirrus cohort. Longitudinally (follow-up up to 3 years), MOGAD-ON with ON within the last 6-12 months before baseline exhibited greater pRNFL thinning than MOGAD-ON with an ON greater than 12 months ago (p < 0.001). The overall MOGAD cohort did not exhibit faster GCIPL thinning compared with the HC cohort., Interpretation: Our study suggests the absence of attack-independent retinal damage in patients with MOGAD. Yet, ongoing neuroaxonal damage or edema resolution seems to occur for up to 12 months after ON, which is longer than what has been reported with other ON forms. These findings support that the pathomechanisms underlying optic nerve involvement and the evolution of OCT retinal changes after ON is distinct in patients with MOGAD. ANN NEUROL 2022;92:476-485., (© 2022 The Authors. Annals of Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.)
- Published
- 2022
- Full Text
- View/download PDF
36. PARIS and SPARTA: Finding the Achilles' Heel of SARS-CoV-2.
- Author
-
Simon V, Kota V, Bloomquist RF, Hanley HB, Forgacs D, Pahwa S, Pallikkuth S, Miller LG, Schaenman J, Yeaman MR, Manthei D, Wolf J, Gaur AH, Estepp JH, Srivastava K, Carreño JM, Cuevas F, Ellebedy AH, Gordon A, Valdez R, Cobey S, Reed EF, Kolhe R, Thomas PG, Schultz-Cherry S, Ross TM, and Krammer F
- Subjects
- COVID-19 Vaccines, Humans, Reinfection, Seroepidemiologic Studies, COVID-19, SARS-CoV-2
- Abstract
To understand reinfection rates and correlates of protection for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we established eight different longitudinal cohorts in 2020 under the umbrella of the PARIS (Protection Associated with Rapid Immunity to SARS-CoV-2)/SPARTA (SARS SeroPrevalence And Respiratory Tract Assessment) studies. Here, we describe the PARIS/SPARTA cohorts, the harmonized assays and analysis that are performed across the cohorts, as well as case definitions for SARS-CoV-2 infection and reinfection that have been established by the team of PARIS/SPARTA investigators. IMPORTANCE Determining reinfection rates and correlates of protection against SARS-CoV-2 infection induced by both natural infection and vaccination is of high significance for the prevention and control of coronavirus disease 2019 (COVID-19). Furthermore, understanding reinfections or infection after vaccination and the role immune escape plays in these scenarios will inform the need for updates of the current SARS-CoV-2 vaccines and help update guidelines suitable for the postpandemic world.
- Published
- 2022
- Full Text
- View/download PDF
37. Impacts of NaHCO 3 on β-Lactam Binding to PBP2a Protein Variants Associated with the NaHCO 3 -Responsive versus NaHCO 3 -Non-Responsive Phenotypes.
- Author
-
Ersoy SC, Chan LC, Yeaman MR, Chambers HF, Proctor RA, Ludwig KC, Schneider T, Manna AC, Cheung A, and Bayer AS
- Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) regulates resistance to β-lactams via preferential production of an alternative penicillin-binding protein (PBP), PBP2a. PBP2a binds many β-lactam antibiotics with less affinity than PBPs which are predominant in methicillin-susceptible (MSSA) strains. A novel, rather frequent in vitro phenotype was recently identified among clinical MRSA bloodstream isolates, termed "NaHCO
3 -responsiveness". This phenotype features β-lactam susceptibility of certain MRSA strains only in the presence of NaHCO3 . Two distinct PBP2a variants, 246G and 246E, have been linked to the NaHCO3 -responsive and NaHCO3 -non-responsive MRSA phenotypes, respectively. To determine the mechanistic impact of PBP2a variants on β-lactam susceptibility, binding profiles of a fluorescent penicillin probe (Bocillin-FL) to each purified PBP2a variant were assessed and compared to whole-cell binding profiles characterized by flow cytometry in the presence vs. absence of NaHCO3 . These investigations revealed that NaHCO3 differentially influenced the binding of the fluorescent penicillin, Bocillin-FL, to the PBP2a variants, with binding intensity and rate of binding significantly enhanced in the 246G compared to the 246E variant. Of note, the NaHCO3 -β-lactam (oxacillin)-responsive JE2 strain, which natively harbors the 246G variant, had enhanced Bocillin-FL whole-cell binding following exposure to NaHCO3 . This NaHCO3 -mediated increase in whole-cell Bocillin-FL binding was not observed in the NaHCO3 -non-responsive parental strain, COL, which contains the 246E PBP2a variant. Surprisingly, genetic swaps of the mecA coding sites between JE2 and COL did not alter the NaHCO3 -enhanced binding seen in JE2 vs. COL. These data suggest that the non-coding regions of mecA may be involved in NaHCO3 -responsiveness. This investigation also provides strong evidence that the NaHCO3 -responsive phenotype in MRSA may involve NaHCO3 -mediated increases in both initial cell surface β-lactam binding, as well as ultimate PBP2a binding of β-lactams.- Published
- 2022
- Full Text
- View/download PDF
38. Mathematical models to study the biology of pathogens and the infectious diseases they cause.
- Author
-
Xavier JB, Monk JM, Poudel S, Norsigian CJ, Sastry AV, Liao C, Bento J, Suchard MA, Arrieta-Ortiz ML, Peterson EJR, Baliga NS, Stoeger T, Ruffin F, Richardson RAK, Gao CA, Horvath TD, Haag AM, Wu Q, Savidge T, and Yeaman MR
- Abstract
Mathematical models have many applications in infectious diseases: epidemiologists use them to forecast outbreaks and design containment strategies; systems biologists use them to study complex processes sustaining pathogens, from the metabolic networks empowering microbial cells to ecological networks in the microbiome that protects its host. Here, we (1) review important models relevant to infectious diseases, (2) draw parallels among models ranging widely in scale. We end by discussing a minimal set of information for a model to promote its use by others and to enable predictions that help us better fight pathogens and the diseases they cause., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
39. Astrocytic outer retinal layer thinning is not a feature in AQP4-IgG seropositive neuromyelitis optica spectrum disorders.
- Author
-
Lu A, Zimmermann HG, Specovius S, Motamedi S, Chien C, Bereuter C, Lana-Peixoto MA, Fontenelle MA, Ashtari F, Kafieh R, Dehghani A, Pourazizi M, Pandit L, D'Cunha A, Kim HJ, Hyun JW, Jung SK, Leocani L, Pisa M, Radaelli M, Siritho S, May EF, Tongco C, De Sèze J, Senger T, Palace J, Roca-Fernández A, Leite MI, Sharma SM, Stiebel-Kalish H, Asgari N, Soelberg KK, Martinez-Lapiscina EH, Havla J, Mao-Draayer Y, Rimler Z, Reid A, Marignier R, Cobo-Calvo A, Altintas A, Tanriverdi U, Yildirim R, Aktas O, Ringelstein M, Albrecht P, Tavares IM, Bichuetti DB, Jacob A, Huda S, Soto de Castillo I, Petzold A, Green AJ, Yeaman MR, Smith TJ, Cook L, Paul F, Brandt AU, and Oertel FC
- Subjects
- Adult, Astrocytes pathology, Autoantibodies, Cross-Sectional Studies, Female, Humans, Male, Middle Aged, Tomography, Optical Coherence, Aquaporin 4 blood, Neuromyelitis Optica physiopathology, Retina physiopathology
- Abstract
Background: Patients with anti-aquaporin-4 antibody seropositive (AQP4-IgG+) neuromyelitis optica spectrum disorders (NMOSDs) frequently suffer from optic neuritis (ON) leading to severe retinal neuroaxonal damage. Further, the relationship of this retinal damage to a primary astrocytopathy in NMOSD is uncertain. Primary astrocytopathy has been suggested to cause ON-independent retinal damage and contribute to changes particularly in the outer plexiform layer (OPL) and outer nuclear layer (ONL), as reported in some earlier studies. However, these were limited in their sample size and contradictory as to the localisation. This study assesses outer retinal layer changes using optical coherence tomography (OCT) in a multicentre cross-sectional cohort., Method: 197 patients who were AQP4-IgG+ and 32 myelin-oligodendrocyte-glycoprotein antibody seropositive (MOG-IgG+) patients were enrolled in this study along with 75 healthy controls. Participants underwent neurological examination and OCT with central postprocessing conducted at a single site., Results: No significant thinning of OPL (25.02±2.03 µm) or ONL (61.63±7.04 µm) were observed in patients who were AQP4-IgG+ compared with patients who were MOG-IgG+ with comparable neuroaxonal damage (OPL: 25.10±2.00 µm; ONL: 64.71±7.87 µm) or healthy controls (OPL: 24.58±1.64 µm; ONL: 63.59±5.78 µm). Eyes of patients who were AQP4-IgG+ (19.84±5.09 µm, p=0.027) and MOG-IgG+ (19.82±4.78 µm, p=0.004) with a history of ON showed parafoveal OPL thinning compared with healthy controls (20.99±5.14 µm); this was not observed elsewhere., Conclusion: The results suggest that outer retinal layer loss is not a consistent component of retinal astrocytic damage in AQP4-IgG+ NMOSD. Longitudinal studies are necessary to determine if OPL and ONL are damaged in late disease due to retrograde trans-synaptic axonal degeneration and whether outer retinal dysfunction occurs despite any measurable structural correlates., Competing Interests: Competing interests: HZ reports grants from Novartis and speaking honoraria from Bayer Healthcare, unrelated to this study. EHM-L received funding from the Instituto de Salud Carlos III (Spain) and Fondo Europeo de Desarrollo Regional (FEDER-JR16/00006), Grant for MS Innovation, Fundació Privada Cellex and Marató TV3 Charitable Foundation and is a researcher in the OCTIMS study, an observational study (that involves no specific drugs) to validate SD-OCT as a biomarker for MS, sponsored by Novartis and has received honoraria and travel support for international and national meetings over the last 3 years from from Biogen, Novartis, Roche, Genzyme. She is a member of the working committee of International Multiple Sclerosis Visual System (IMSVISUAL) Consortium. MAL-P has received funding for travel and speaker honoraria from Novartis, Sanofi- Genzyme and Roche. MAF has nothing to disclose. Jacqueline Palace has received support for scientific meetings and honorariums for advisory work From Merck Serono, Novartis, Chugai, Alexion, Roche, Medimmune, Argenx, UCB, Mitsubishi, Amplo, Janssen. Grants from Alexion, Amplo biotechnology. Shares in AstraZenica. Acknowledges Partial funding by Highly specialised services NHS England. MIL reported being involved in aquaporin 4 testing, receiving salary from the National Health Service National Highly Specialised Commissioning Group for Neuromyelitis Optica, UK, being supported by the National Institute for Health Research Oxford Biomedical Research Centre, UK, and receiving speaking honoraria and travel grants from Biogen Idec, and travel grant from Novartis. SMS has nothing to disclose. AR-F is sponsored by Abide Therapeutic outside of the submitted work and reports no potential conflicts of interest. SSiritho received funding for travel and speaker honoraria from Merck Serono, Pacific Healthcare (Thailand), Menarini (Thailand), Biogen Idec, UCB (Thailand), and Novartis. AA reports personal fees from received honoraria for giving educational presentations on multiple sclerosis and neuroimmunology at several national congresses or symposia from Teva Turkey, Merck-Serono, Biogen Idec-Gen Pharma of Turkey, Roche, Novartis, Bayer, Sanofi-Genzyme. She has received travel and registration coverage for attending several national and international congresses or symposia from Merck-Serono, Biogen Idec-Gen Pharma of Turkey, Roche, Sanofi-Genzyme and Bayer. AJ has received compensation for advisory board, consulting, meeting attendance and speaking from Biogen, Terumo-BCT, Genentech, Shire and Chugai Pharmaceuticals. SH has received funding from the NMO Spectrum-UK charity and was previously funded by an MGA/Watney/NIHR Oxford Biomedical research grant. RM serves on scientific advisory board for MedImmune and has received funding for travel and honoraria from Biogen, Merck Serono, Novartis, Sanofi- Genzyme, Roche and Teva. EN has nothing to disclose. ACC received funding from the Instituto de Salud Carlos III (Spain) JR19/00007 unrelated to this manuscript. DB has received speaking/consulting honoraria from Bayer Health Care, Biogen Idec, Merck, Sanofi-Genzyme, TEVA and Roche and had travel expenses to scientific meetings sponsored by Bayer Health Care, Merck Serono, TEVA and Roche. JH reports grants for OCT research from the Friedrich-Baur-Stiftung and Merck, personal fees and non-financial support from Celgene, Merck, Alexion, Novartis, Roche, Santhera, Biogen, Heidelberg Engineering, Sanofi Genzyme and non-financial support of the Guthy-Jackson Charitable Foundation, all outside the submitted work. JH is partially funded by the German Federal Ministry of Education and Research (DIFUTURE), Grant Numbers 01ZZ1603[A-D] and 01ZZ1804[A-H]. LL received honoraria for consulting services from Merck, Roche, Biogen and for speaking activities from Teva; research support from Merck, Biogen, Novartis; travel support from Merck, Roche, Biogen, Almirall. MP has nothing to disclose. OA has received honoraria for speaking/consultation and travel grants from Bayer Healthcare, Biogen Idec, Chugai, Novartis, Medimmune, Merck Serono, and Teva and research grants from Bayer Healthcare, Biogen Idec, Novartis, and Teva. MR received speaker honoraria from Novartis, Bayer, Roche, Alexion and Ipsen and travel reimbursement from Bayer, Biogen, Merz, Genzyme, Teva, Roche and Merck, none related to this study. PA reports grants, personal fees and non-financial support from Allergan, Biogen, Ipsen, Merz Pharmaceuticals, Novartis, and Roche, personal fees and non-financial support from Bayer Healthcare, and Merck, and non-financial support from Sanofi-Aventis/Genzyme. HJK reports speaking and/or consulting: Bayer Schering Pharma, Biogen, Celltrion, Eisai, HanAll BioPharma, MedImmune, Merck Serono, Novartis, Sanofi Genzyme, Teva-Handok, and UCB; research support: Ministry of Science & ICT, Sanofi Genzyme, Teva-Handok, and UCB; steering committee member: MedImmune; co-editor/associated editor: MS Journal-Experimental, Translational and Clinical; and Journal of Clinical Neurology. J-WH has received a grant from the National Research Foundation of Korea. YM-D has served as a consultant and/or received grant support from: Acorda, Bayer Pharmaceutical, Biogen Idec, Celgene, EMD Serono, Genzyme, Novartis, Questor, Chugai, and Teva Neuroscience and is currently supported by grants from NIH NIAID Autoimmune Center of Excellence: UM1-AI110557; NIH NINDS R01-NS080821. HSK has nothing to disclose. IK served on scientific advisory board for Biogen Idec and Genentech and received research support from Guthy-Jackson Charitable Foundation, National Multiple Sclerosis Society, Biogen-Idec,Serono, Genzyme and Novartis. ZR has nothing to disclose. AR has nothing to disclose. MRY is founder and a shareholder of NovaDigm Therapeutics, Inc; he receives funding from the United States National Institutes of Health and United States Department of Defense; he holds US and international patents on immunotherapeutic and anti-infective technologies, is a member of the Genentech-Roche Scientific Advisory Committee and adviser to The Guthy-Jackson Charitable Foundation. TJS was issued US patents covering the therapeutic targeting of IGF-I receptor in autoimmune diseases. He is a paid consultant for Horizon Thera and Immunovant and is a scientific advisor to the Guthy-Jackson Charitable Foundation. He receives research funding from the National Institutes of Health. AP is supported by the National Institute for Health Research (NIHR) Biomedical Research Centre based at Moorfields Eye Hospital National Health Service (NHS) Foundation Trust and University College London Institute of Ophthalmology. AB is cofounder and shareholder of Motognosis and Nocturne. He is named as inventor on several patent applications regarding MS serum biomarkers, OCT image analysis and perceptive visual computing. FP reports research grants and speaker honoraria from Bayer, Teva, Genzyme, Merck, Novartis, MedImmune and is member of the steering committee of the OCTIMS study (Novartis), all unrelated to this work. FCO was employee of Nocturne GmbH and receives research support by the American Academy of Neurology and National Multiple Sclerosis Society (US), unrelated to this work as well as funding by the German Association of Neurology (Deutsche Gesellschaft für Neurologie) in context of this project.CC has received a speaking honorarium from Bayer and research funding from Novartis unrelated to this publication. All other authors have nothing to disclose., (© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2022
- Full Text
- View/download PDF
40. New Mechanistic Insights into Purine Biosynthesis with Second Messenger c-di-AMP in Relation to Biofilm-Related Persistent Methicillin-Resistant Staphylococcus aureus Infections.
- Author
-
Li L, Li Y, Zhu F, Cheung AL, Wang G, Bai G, Proctor RA, Yeaman MR, Bayer AS, and Xiong YQ
- Subjects
- Anti-Bacterial Agents pharmacology, Bacterial Proteins genetics, Bacterial Proteins metabolism, Biofilms drug effects, Biosynthetic Pathways, Daptomycin pharmacology, Humans, Methicillin-Resistant Staphylococcus aureus drug effects, Methicillin-Resistant Staphylococcus aureus genetics, Second Messenger Systems, Cyclic AMP metabolism, Methicillin-Resistant Staphylococcus aureus metabolism, Persistent Infection microbiology, Purines biosynthesis, Staphylococcal Infections microbiology
- Abstract
Persistent methicillin-resistant Staphylococcus aureus (MRSA) endovascular infections represent a significant clinically challenging subset of invasive, life-threatening S. aureus infections. We have recently demonstrated that purine biosynthesis plays an important role in such persistent infections. Cyclic di-AMP (c-di-AMP) is an essential and ubiquitous second messenger that regulates many cellular pathways in bacteria. However, whether there is a regulatory connection between the purine biosynthesis pathway and c-di-AMP impacting persistent outcomes was not known. Here, we demonstrated that the purine biosynthesis mutant MRSA strain, the Δ purF strain (compared to its isogenic parental strain), exhibited the following significant differences in vitro : (i) lower ADP, ATP, and c-di-AMP levels; (ii) less biofilm formation with decreased extracellular DNA (eDNA) levels and Triton X-100-induced autolysis paralleling enhanced expressions of the biofilm formation-related two-component regulatory system lytSR and its downstream gene lrgB ; (iii) increased vancomycin (VAN)-binding and VAN-induced lysis; and (iv) decreased wall teichoic acid (WTA) levels and expression of the WTA biosynthesis-related gene, tarH . Substantiating these data, the dacA (encoding diadenylate cyclase enzyme required for c-di-AMP synthesis) mutant strain ( dacA
G206S strain versus its isogenic wild-type MRSA and dacA -complemented strains) showed significantly decreased c-di-AMP levels, similar in vitro effects as seen above for the purF mutant and hypersusceptible to VAN treatment in an experimental biofilm-related MRSA endovascular infection model. These results reveal an important intersection between purine biosynthesis and c-di-AMP that contributes to biofilm-associated persistence in MRSA endovascular infections. This signaling pathway represents a logical therapeutic target against persistent MRSA infections. IMPORTANCE Persistent endovascular infections caused by MRSA, including vascular graft infection syndromes and infective endocarditis, are significant and growing public health threats. A particularly worrisome trend is that most MRSA isolates from these patients are "susceptible" in vitro to conventional anti-MRSA antibiotics, such as VAN and daptomycin (DAP), based on Clinical and Laboratory Standards Institute breakpoints. Yet, these antibiotics frequently fail to eliminate these infections in vivo . Therefore, the persistent outcomes in MRSA infections represent a unique and important variant of classic "antibiotic resistance" that is only disclosed during in vivo antibiotic treatment. Given the high morbidity and mortality associated with the persistent infection, there is an urgent need to understand the specific mechanism(s) of this syndrome. In the current study, we demonstrate that a functional intersection between purine biosynthesis and the second messenger c-di-AMP plays an important role in VAN persistence in experimental MRSA endocarditis. Targeting this pathway may represent a potentially novel and effective strategy for treating these life-threatening infections.- Published
- 2021
- Full Text
- View/download PDF
41. Cytoprotective IgG antibodies in sera from a subset of patients with AQP4-IgG seropositive neuromyelitis optica spectrum disorder.
- Author
-
Tradtrantip L, Yeaman MR, and Verkman AS
- Subjects
- Animals, Aquaporin 4 blood, Autoantibodies blood, Autoantibodies immunology, Biomarkers blood, CHO Cells, Cricetulus, Disease Progression, Humans, Immune Sera, Immunoglobulin G blood, Neuromyelitis Optica blood, Neuromyelitis Optica pathology, Aquaporin 4 immunology, Neuromyelitis Optica immunology
- Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune inflammatory disease of the central nervous system. Most NMOSD patients are seropositive for immunoglobulin G (IgG) autoantibodies against astrocyte water channel aquaporin-4 (AQP4), called AQP4-IgG. AQP4-IgG binding to aquaporin-4 causes complement-dependent cytotoxicity (CDC), leading to inflammation and demyelination. Here, CDC was measured in AQP4-expressing cells exposed to human complement and heat-inactivated sera from 108 AQP4-IgG seropositive NMOSD subjects and 25 non-NMOSD controls. AQP4-IgG positive sera produced a wide range of CDC, with 50% maximum cytotoxicity produced by as low as 0.2% serum concentration. Unexpectedly, 58 samples produced no cytotoxicity, and of those, four sera were cytoprotective against cytotoxic AQP4-IgG. Cytoprotection was found against different cytotoxic monoclonal AQP4-IgGs and NMOSD patient sera, and in primary astrocyte cultures. Mechanistic studies revealed that the protective factor is an IgG antibody that did not inhibit complement directly, but interfered with binding of cytotoxic AQP4-IgG to AQP4 and consequent C1q binding and complement activation. Further studies suggested that non-pathogenic AQP4-IgG, perhaps with altered glycosylation, may contribute to reduced or ineffectual binding of cytotoxic AQP4-IgG, as well as reduced cell-surface AQP4. The presence of natural cytoprotective antibodies in AQP4-IgG seropositive sera reveals an added level of complexity in NMOSD disease pathogenesis, and suggests the potential therapeutic utility of 'convalescent' serum or engineered protective antibody to interfere with pathogenic antibody in AQP4-IgG seropositive NMOSD., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
42. Role of the Staphylococcus aureus Extracellular Loop of GraS in Resistance to Distinct Human Defense Peptides in PMN and Invasive Cardiovascular infections.
- Author
-
Cheung AL, Cho J, Bayer AS, Yeaman MR, Xiong YQ, Donegan NP, Mikheyeva IV, Lee GY, and Yang SJ
- Subjects
- Animals, Antimicrobial Cationic Peptides metabolism, Endocarditis metabolism, Endocarditis microbiology, Female, Gene Expression Regulation, Bacterial genetics, Humans, Microbial Sensitivity Tests methods, Microbial Viability genetics, Neutrophils microbiology, Rabbits, Staphylococcal Infections microbiology, Bacterial Proteins genetics, Cardiovascular Infections metabolism, Cardiovascular Infections microbiology, Drug Resistance, Bacterial genetics, Methicillin-Resistant Staphylococcus aureus genetics, Neutrophils metabolism, Staphylococcal Infections metabolism
- Abstract
GraS is a membrane sensor in Staphylococcus aureus that induces mprF and dltABCD expression to alter the surface positive charge upon exposure to cationic human defense peptides (HDPs). The sensing domain of GraS likely resides in the 9-residue extracellular loop (EL). In this study, we assessed a hospital-acquired methicillin-resistant S. aureus (HA-MRSA) strain (COL) for the specific role of two distinct EL mutations: F38G (bulk) and D/35/37/41K (charged inversion). Activation of mprF by polymyxin B (PMB) was reduced in the D35/37/41K mutant versus the D35/37/41G mutant, correlating with reduced surface positive charge; in contrast, these effects were less prominent in the F38G mutant but still lower than those in the parent. These data indicated that both electrostatic charge and steric bulk of the EL of GraS influence induction of genes impacting HDP resistance. Using mprF expression as a readout, we confirmed GraS signaling was pH dependent, increasing as pH was lowered (from pH 7.5 down to pH 5.5). In contrast to PMB activation, reduction of mprF was comparable at pH 5.5 between the P38G and D35/37/41K point mutants, indicating a mechanistic divergence between GraS activation by acidic pH versus cationic peptides. Survival assays in human blood and purified polymorphonuclear leukocytes (PMNs) revealed lower survival of the D35/37/41K mutant versus the F38G mutant, with both being lower than that of the parent. Virulence studies in the rabbit endocarditis model mirrored whole blood and PMN killing assay data described above. Collectively, these data confirmed the importance of specific residues within the EL of GraS in conferring essential bacterial responses for MRSA survival in infections.
- Published
- 2021
- Full Text
- View/download PDF
43. Retinal Optical Coherence Tomography in Neuromyelitis Optica.
- Author
-
Oertel FC, Specovius S, Zimmermann HG, Chien C, Motamedi S, Bereuter C, Cook L, Lana Peixoto MA, Fontanelle MA, Kim HJ, Hyun JW, Palace J, Roca-Fernandez A, Leite MI, Sharma S, Ashtari F, Kafieh R, Dehghani A, Pourazizi M, Pandit L, D'Cunha A, Aktas O, Ringelstein M, Albrecht P, May E, Tongco C, Leocani L, Pisa M, Radaelli M, Martinez-Lapiscina EH, Stiebel-Kalish H, Siritho S, de Seze J, Senger T, Havla J, Marignier R, Cobo-Calvo A, Bichuetti D, Tavares IM, Asgari N, Soelberg K, Altintas A, Yildirim R, Tanriverdi U, Jacob A, Huda S, Rimler Z, Reid A, Mao-Draayer Y, Soto de Castillo I, Petzold A, Green AJ, Yeaman MR, Smith T, Brandt AU, and Paul F
- Subjects
- Adult, Cross-Sectional Studies, Female, Humans, Male, Middle Aged, Neuromyelitis Optica diagnostic imaging, Optic Neuritis diagnostic imaging, Retrospective Studies, Tomography, Optical Coherence, Young Adult, Aquaporin 4 immunology, Neuromyelitis Optica immunology, Neuromyelitis Optica pathology, Optic Neuritis immunology, Optic Neuritis pathology, Retinal Neurons pathology
- Abstract
Background and Objectives: To determine optic nerve and retinal damage in aquaporin-4 antibody (AQP4-IgG)-seropositive neuromyelitis optica spectrum disorders (NMOSD) in a large international cohort after previous studies have been limited by small and heterogeneous cohorts., Methods: The cross-sectional Collaborative Retrospective Study on retinal optical coherence tomography (OCT) in neuromyelitis optica collected retrospective data from 22 centers. Of 653 screened participants, we included 283 AQP4-IgG-seropositive patients with NMOSD and 72 healthy controls (HCs). Participants underwent OCT with central reading including quality control and intraretinal segmentation. The primary outcome was thickness of combined ganglion cell and inner plexiform (GCIP) layer; secondary outcomes were thickness of peripapillary retinal nerve fiber layer (pRNFL) and visual acuity (VA)., Results: Eyes with ON (NMOSD-ON, N = 260) or without ON (NMOSD-NON, N = 241) were assessed compared with HCs (N = 136). In NMOSD-ON, GCIP layer (57.4 ± 12.2 μm) was reduced compared with HC (GCIP layer: 81.4 ± 5.7 μm, p < 0.001). GCIP layer loss (-22.7 μm) after the first ON was higher than after the next (-3.5 μm) and subsequent episodes. pRNFL observations were similar. NMOSD-NON exhibited reduced GCIP layer but not pRNFL compared with HC. VA was greatly reduced in NMOSD-ON compared with HC eyes, but did not differ between NMOSD-NON and HC., Discussion: Our results emphasize that attack prevention is key to avoid severe neuroaxonal damage and vision loss caused by ON in NMOSD. Therapies ameliorating attack-related damage, especially during a first attack, are an unmet clinical need. Mild signs of neuroaxonal changes without apparent vision loss in ON-unaffected eyes might be solely due to contralateral ON attacks and do not suggest clinically relevant progression but need further investigation., (Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.)
- Published
- 2021
- Full Text
- View/download PDF
44. Balancing Potential Benefits and Risks of Bruton Tyrosine Kinase Inhibitor Therapies in Multiple Sclerosis During the COVID-19 Pandemic.
- Author
-
Weber MS, Nicholas JA, and Yeaman MR
- Subjects
- Adult, COVID-19 Vaccines, Humans, Multiple Sclerosis immunology, Prospective Studies, T-Lymphocytes immunology, Agammaglobulinaemia Tyrosine Kinase antagonists & inhibitors, COVID-19 immunology, Multiple Sclerosis drug therapy, Pandemics, Protein Kinase Inhibitors therapeutic use
- Abstract
Bruton tyrosine kinase inhibitors (BTKis) encompass a new class of therapeutics currently being evaluated for the treatment of multiple sclerosis (MS). Whether BTKis affect COVID-19 risk or severity or reduce vaccine efficacy are important but unanswered questions. Here, we provide an overview on BTKi mechanisms relevant to COVID-19 infection and vaccination and review preliminary data on BTKi use in patients with COVID-19. BTKis block B-cell receptor- and myeloid fragment crystallizable receptor-mediated signaling, thereby dampening B-cell activation, antibody class-switching, expansion, and cytokine production. Beyond antibodies, COVID-19 severity and vaccine efficacy appear largely linked to T-cell responses and interferon induction, processes not directly affected by BTKis. Given that B cells have clear roles in antigen presentation to T cells, however, it is possible that BTKis may indirectly interfere with beneficial or detrimental T-cell responses during COVID-19 infection or vaccination. In addition to these possible effects on generating a protective immune response, BTKis may attenuate the hyperinflammatory dysregulation often seen in severe cases of COVID-19 that evolves as a key risk factor in this disease. Currently available outcomes from BTKi-treated patients with COVID-19 are discussed. Clinical trials are currently underway to evaluate the safety and efficacy of BTKis in individuals with MS. Although limited data suggest a potential benefit of BTKis on outcomes for some COVID-19 patients, data from adequately powered, prospective and randomized clinical trials are lacking. Likewise, the specific effect of BTKis on the safety and efficacy of COVID-19 vaccines remains to be determined. Any potential unknown risks that BTKi therapy may present to the patient relative to COVID-19 infection, severity, and vaccine efficacy must be balanced with the importance of timely intervention to prevent or minimize MS progression., (Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.)
- Published
- 2021
- Full Text
- View/download PDF
45. Immunosuppression in Glomerular Diseases: Implications for SARS-CoV-2 Vaccines and COVID-19.
- Author
-
Yeaman MR
- Abstract
Background: Glomerular diseases (GD) are chronic conditions that often involve immune dysfunction and require immunosuppressive therapy (IST) to control underlying pathogenesis. Unfortunately, such diseases appear to heighten risks of severe outcomes in COVID-19 and predispose to other infections that may be life-threatening. Thus, averting preventable infections is imperative in GD patients., Summary: The advent of vaccines demonstrated to be safe and efficacious against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has favorably impacted the COVID-19 pandemic epidemiology. However, patients on ISTs were excluded from initial vaccine clinical trials. Thus, only limited and incomplete data are available currently regarding the potential impact of immunosuppression on immune response to or efficacy of the SARS-CoV-2 vaccines. However, new insights are emerging from SARS-CoV-2 vaccine studies, and impacts of ISTs on conventional vaccines are useful to consider. Mechanisms of immunosuppressive agents commonly used in the treatment of GD are reviewed with respect to implications for immune responses induced by SARS-CoV-2 vaccines. ISTs discussed include corticosteroids; alkylating agents; antimetabolites; calcineurin or mammalian target of rapamycin inhibitors; CD38+, CD20+, or CD19+ cell depletion; and complement protein C5 inhibition., Key Messages: Many immunosuppressive therapies may potentially attenuate or impair protective immunity of the SARS-CoV-2 vaccines. However, as vaccines currently in use employ mRNA or nonreplicative viral vectors, they appear to be safe in patients on immunosuppression, further favoring vaccination. Moreover, predominant SARS-CoV-2 vaccines are likely to afford at least partial protective immunity through one or more immune mechanisms even in patients on IST. Guidelines and emerging strategies are also considered to optimize vaccine protection from COVID-19., Competing Interests: The author has no conflicts of interest to declare., (Copyright © 2021 by S. Karger AG, Basel.)
- Published
- 2021
- Full Text
- View/download PDF
46. The costs of care from a US claims database in patients with neuromyelitis optica spectrum disorder.
- Author
-
Exuzides A, Sheinson D, Sidiropoulos P, Gholizadeh S, Magrini F, Surinach A, Cook L, Meyer CS, and Yeaman MR
- Subjects
- Aged, Ambulatory Care, Databases, Factual, Hospitalization, Humans, Medicare, United States, Neuromyelitis Optica therapy
- Abstract
Background: Neuromyelitis optica spectrum disorder (NMOSD) is a rare autoimmune disease of the central nervous system that often leads to severe disability. Patients with highly active NMOSD have approximately a 10-times higher hospital inpatient admission rate compared with patients without NMOSD. Accurate assessments of the impact of NMOSD treatments on the burdens of illness require quantitative metrics of these burdens, including costs of care., Methods: This study evaluated claims data from the IBM MarketScan Commercial and Medicare Supplemental Databases between 2014 and 2018. Patients were included based on inpatient or outpatient claims meeting criteria defined for NMOSD. Non-NMOSD controls were matched 5:1 to patients with NMOSD. Total costs of healthcare services in consumer price index-adjusted 2019 US dollars during the 1-year postindex follow-up period were calculated for patients and controls., Results: Patients with NMOSD required more healthcare services and incurred significantly greater costs for inpatient hospitalizations (annual mean [SD] cost: $29,054 [$144,872] vs controls $1521 [$10,759]), outpatient services ($24,881 [$35,463] vs $4761 [$26,447]), and emergency department (ED) visits ($2400 [$7771] vs $408 [$2579]). Almost 12% of patients with NMOSD were further burdened with plasma exchange or intravenous immunoglobulin G treatments, costing an annual median (interquartile range) of $1684 ($566-$3817) and $24,353 ($5425-$42,975), respectively., Conclusions: Compared with controls, patients with NMOSD had significantly higher costs associated with hospitalizations, ED visits, and prescriptions. These results highlight the considerable economic burden of NMOSD, which may be favorably impacted by disease-modifying therapies that are regulatory-approved to be safe and effective., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
47. Platelet Deficiency Represents a Modifiable Risk Factor for Periprosthetic Joint Infection in a Preclinical Mouse Model.
- Author
-
Greig D, Trikha R, Sekimura T, Cevallos N, Kelley BV, Mamouei Z, Yeaman MR, and Bernthal NM
- Subjects
- Animals, Biofilms, Disease Models, Animal, Mice, Prosthesis-Related Infections microbiology, Risk Factors, Staphylococcal Infections microbiology, Thrombocytopenia microbiology, Prosthesis-Related Infections etiology, Staphylococcal Infections etiology, Staphylococcus aureus isolation & purification, Thrombocytopenia complications
- Abstract
Background: Well known for their hemostatic function, platelets are increasingly becoming recognized as important immunomodulators. The purpose of the present study was to assess the impact of platelet depletion on antimicrobial host defense in a mouse model of periprosthetic joint infection (PJI)., Methods: Thrombocytopenia (TCP) was induced in C57BL/6 mice with use of a selective antibody against platelet CD41 (anti-CD41). Whole blood from pre-treated mice was incubated with Staphylococcus aureus to assess antimicrobial efficacy with use of bioluminescent imaging, quantitative histological staining, and colony forming unit (CFU) quantification. In parallel, untreated heterologous platelets were added to TCP blood to assess potential rescue of antimicrobial efficacy. In vivo, TCP and control mice underwent placement of a titanium implant in the femur inoculated with bioluminescent Xen36 S. aureus. Longitudinal bioluminescent imaging was performed postoperatively to quantify the evolution of bacterial burden, which was confirmed via assessment of S. aureus CFUs on the implant and in peri-implant tissue on postoperative day (POD) 28., Results: Anti-CD41 treatment resulted in significant dose-dependent reductions in platelet count. Ex vivo, platelet-depleted whole blood demonstrated significantly less bacterial reduction than control blood. These outcomes were reversed with the addition of untreated rescue platelets. In vivo, infection burden was significantly higher in TCP mice and was inversely correlated with preoperative platelet count (r2 = 0.63, p = 0.037). Likewise, CFU quantification on POD28 was associated with increased bacterial proliferation and severity of periprosthetic infection in TCP mice compared with controls., Conclusions: Thrombocytopenia resulted in an increased bacterial burden both ex vivo and in vivo in a mouse model of PJI., Clinical Relevance: In orthopaedic patients, deficiencies in platelet quantity or function represent an easily modifiable risk factor for PJI., Competing Interests: Disclosure: This study was supported by the H&H Lee Research Program and the National Institute of Arthritis and Musculoskeletal and Skin Diseases of the National Institutes of Health. On the Disclosure of Potential Conflicts of Interest forms, which are provided with the online version of the article, one or more of the authors checked “yes” to indicate that the author had a relevant financial relationship in the biomedical arena outside the submitted work (http://links.lww.com/JBJS/G396)., (Copyright © 2021 by The Journal of Bone and Joint Surgery, Incorporated.)
- Published
- 2021
- Full Text
- View/download PDF
48. Mechanistic Fingerprinting Reveals Kinetic Signatures of Resistance to Daptomycin and Host Defense Peptides in Streptococcus mitis-oralis .
- Author
-
Yeaman MR, Chan LC, Mishra NN, and Bayer AS
- Abstract
Streptococcus mitis-oralis (S. mitis-oralis) infections are increasingly prevalent in specific populations, including neutropenic cancer and endocarditis patients. S. mitis-oralis strains have a propensity to evolve rapid, high-level and durable resistance to daptomycin (DAP-R) in vitro and in vivo, although the mechanism(s) involved remain incompletely defined. We examined mechanisms of DAP-R versus cross-resistance to cationic host defense peptides (HDPs), using an isogenic S. mitis-oralis strain-pair: (i) DAP-susceptible (DAP-S) parental 351-WT (DAP MIC = 0.5 µg/mL), and its (ii) DAP-R variant 351-D10 (DAP MIC > 256 µg/mL). DAP binding was quantified by flow cytometry, in-parallel with temporal (1-4 h) killing by either DAP or comparative prototypic cationic HDPs (hNP-1; LL-37). Multicolor flow cytometry was used to determine kinetic cell responses associated with resistance or susceptibility to these molecules. While overall DAP binding was similar between strains, a significant subpopulation of 351-D10 cells hyper-accumulated DAP (>2-4-fold vs. 351-WT). Further, both DAP and hNP-1 induced cell membrane (CM) hyper-polarization in 351-WT, corresponding to significantly greater temporal DAP-killing (vs. 351-D10). No strain-specific differences in CM permeabilization, lipid turnover or regulated cell death were observed post-exposure to DAP, hNP-1 or LL-37. Thus, the adaptive energetics of the CM appear coupled to the outcomes of interactions of S. mitis-oralis with DAP and selected HDPs. In contrast, altered CM permeabilization, proposed as a major mechanism of action of both DAP and HDPs, did not differentiate DAP-S vs. DAP-R phenotypes in this S. mitis-oralis strain-pair.
- Published
- 2021
- Full Text
- View/download PDF
49. Human DNA methylation signatures differentiate persistent from resolving MRSA bacteremia.
- Author
-
Chang YL, Rossetti M, Gjertson DW, Rubbi L, Thompson M, Montoya DJ, Morselli M, Ruffin F, Hoffmann A, Pellegrini M, Fowler VG Jr, Yeaman MR, and Reed EF
- Subjects
- Anti-Bacterial Agents administration & dosage, Bacteremia drug therapy, CCAAT-Enhancer-Binding Protein-beta metabolism, Female, Humans, Male, Middle Aged, STAT1 Transcription Factor metabolism, Staphylococcal Infections drug therapy, p300-CBP Transcription Factors metabolism, Bacteremia metabolism, DNA Methylation, Methicillin-Resistant Staphylococcus aureus metabolism, Response Elements, Staphylococcal Infections metabolism
- Abstract
Persistent methicillin-resistant Staphylococcus aureus (MRSA) bacteremia is life threatening and occurs in up to 30% of MRSA bacteremia cases despite appropriate antimicrobial therapy. Isolates of MRSA that cause antibiotic-persistent methicillin-resistant S. aureus bacteremia (APMB) typically have in vitro antibiotic susceptibilities equivalent to those causing antibiotic-resolving methicillin-resistant S. aureus bacteremia (ARMB). Thus, persistence reflects host-pathogen interactions occurring uniquely in context of antibiotic therapy in vivo. However, host factors and mechanisms involved in APMB remain unclear. We compared DNA methylomes in circulating immune cells from patients experiencing APMB vs. ARMB. Overall, methylation signatures diverged in the distinct patient cohorts. Differentially methylated sites intensified proximate to transcription factor binding sites, primarily in enhancer regions. In APMB patients, significant hypomethylation was observed in binding sites for CCAAT enhancer binding protein-β (C/EBPβ) and signal transducer/activator of transcription 1 (STAT1). In contrast, hypomethylation in ARMB patients localized to glucocorticoid receptor and histone acetyltransferase p300 binding sites. These distinct methylation signatures were enriched in neutrophils and achieved a mean area under the curve of 0.85 when used to predict APMB using a classification model. These findings validated by targeted bisulfite sequencing (TBS-seq) differentiate epigenotypes in patients experiencing APMB vs. ARMB and suggest a risk stratification strategy for antibiotic persistence in patients treated for MRSA bacteremia., Competing Interests: Competing interest statement: V.G.F. reports grant/research support from MedImmune, Cerexa/Forest/Actavis/Allergan, Pfizer, Advanced Liquid Logics, Theravance, Novartis, Cubist/Merck, Medical Biosurfaces, Locus, Affinergy, Contrafect, Karius, Genentech, Regeneron, and Basilea; paid consultant for Pfizer, Novartis, Galderma, Novadigm, Durata, Debiopharm, Genentech, Achaogen, Affinium, Medicines Co., Cerexa, Tetraphase, Trius, MedImmune, Bayer, Theravance, Cubist, Basilea, Affinergy, Janssen, xBiotech, Contrafect, Regeneron, Basilea, and Destiny; membership with Merck as cochair for V710 vaccine; educational fees from Green Cross, Cubist, Cerexa, Durata, Theravance, and Debiopharm; and royalties from UpToDate. M.R.Y. is a founder and shareholder of NovaDigm Therapeutics, Inc., which develops vaccines and immunotherapeutics targeting multidrug-resistant pathogens, including S. aureus.
- Published
- 2021
- Full Text
- View/download PDF
50. Identification of Candida glabrata Transcriptional Regulators That Govern Stress Resistance and Virulence.
- Author
-
Filler EE, Liu Y, Solis NV, Wang L, Diaz LF, Edwards JE Jr, Filler SG, and Yeaman MR
- Subjects
- Gene Deletion, Genetic Variation, Humans, Mutation, Candida glabrata genetics, Candida glabrata pathogenicity, Fungal Proteins genetics, Host-Pathogen Interactions genetics, Transcription Factors genetics, Virulence genetics
- Abstract
The mechanisms by which Candida glabrata resists host defense peptides and caspofungin are incompletely understood. To identify transcriptional regulators that enable C. glabrata to withstand these classes of stressors, a library of 215 C. glabrata transcriptional regulatory deletion mutants was screened for susceptibility to both protamine and caspofungin. We identified eight mutants that had increased susceptibility to both host defense peptides and caspofungin. Of these mutants, six were deleted for genes that were predicted to specify proteins involved in histone modification. These genes were ADA2 , GCN5 , SPT8 , HOS2 , RPD3 , and SPP1 Deletion of ADA2 , GCN5 , and RPD3 also increased susceptibility to mammalian host defense peptides. The Δ ada2 and Δ gcn5 mutants had increased susceptibility to other stressors, such as H
2 O2 and SDS. In the Galleria mellonella model of disseminated infection, the Δ ada2 and Δ gcn5 mutants had attenuated virulence, whereas in neutropenic mice, the virulence of the Δ ada2 and Δ rpd3 mutants was decreased. Thus, histone modification plays a central role in enabling C. glabrata to survive host defense peptides and caspofungin, and Ada2 and Rpd3 are essential for the maximal virulence of this organism during disseminated infection., (Copyright © 2021 American Society for Microbiology.)- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.