1
|
Kim JK, Yun HY, Kim JS, Kim W, Lee CS, Kim BG, Jeong HJ. Development of fluorescence-linked immunosorbent assay for rapid detection of Staphylococcus aureus. Appl Microbiol Biotechnol 2024; 108:2. [PMID: 38153552 DOI: 10.1007/s00253-023-12836-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/22/2023] [Accepted: 09/30/2023] [Indexed: 12/29/2023]
Abstract
Staphylococcus aureus is a major pathogen that causes infections and life-threatening diseases. Although antibiotics, such as methicillin, have been used, methicillin-resistant S. aureus (MRSA) causes high morbidity and mortality rates, and conventional detection methods are difficult to be used because of time-consuming process. To control the spread of S. aureus, a development of a rapid and simple detection method is required. In this study, we generated a fluorescent anti-S. aureus antibody, and established a novel fluorescence-linked immunosorbent assay (FLISA)-based S. aureus detection method. The method showed high sensitivity and low limit of detection toward MRSA detection. The assay time for FLISA was 5 h, which was faster than that of conventional enzyme-linked immunosorbent assay (ELISA) or rapid ELISA. Moreover, the FLISA-based detection method was applied to diagnose clinically isolated MRSA samples that required only 5.3 h of preincubation. The FLISA method developed in this study can be widely applied as a useful tool for convenient S. aureus detection. KEY POINTS: • A fluorescence-linked immunosorbent assay-based S. aureus detection method • Simultaneous quantification of a maximum of 96 samples within 5 h • Application of the novel system to diagnosis clinical isolates.
Collapse
Affiliation(s)
- Joo-Kyung Kim
- Interdisciplinary Program for Biochemical Engineering and Biotechnology, Seoul National University, Seoul, 08826, South Korea
| | - Hyun-Young Yun
- Department of Biological and Chemical Engineering, Hongik University, Sejong, 30016, South Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, 05355, South Korea
| | - Wooseong Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Chang-Soo Lee
- Department of Chemical Engineering and Applied Chemistry, Chungnam National University, Daejeon, 34134, South Korea
| | - Byung-Gee Kim
- Interdisciplinary Program for Biochemical Engineering and Biotechnology, Seoul National University, Seoul, 08826, South Korea
| | - Hee-Jin Jeong
- Department of Biological and Chemical Engineering, Hongik University, Sejong, 30016, South Korea.
| |
Collapse
|
2
|
Holmes CL, Albin OR, Mobley HLT, Bachman MA. Bloodstream infections: mechanisms of pathogenesis and opportunities for intervention. Nat Rev Microbiol 2024:10.1038/s41579-024-01105-2. [PMID: 39420097 DOI: 10.1038/s41579-024-01105-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 10/19/2024]
Abstract
Bloodstream infections (BSIs) are common in hospitals, often life-threatening and increasing in prevalence. Microorganisms in the blood are usually rapidly cleared by the immune system and filtering organs but, in some cases, they can cause an acute infection and trigger sepsis, a systemic response to infection that leads to circulatory collapse, multiorgan dysfunction and death. Most BSIs are caused by bacteria, although fungi also contribute to a substantial portion of cases. Escherichia coli, Staphylococcus aureus, coagulase-negative Staphylococcus, Klebsiella pneumoniae and Candida albicans are leading causes of BSIs, although their prevalence depends on patient demographics and geographical region. Each species is equipped with unique factors that aid in the colonization of initial sites and dissemination and survival in the blood, and these factors represent potential opportunities for interventions. As many pathogens become increasingly resistant to antimicrobials, new approaches to diagnose and treat BSIs at all stages of infection are urgently needed. In this Review, we explore the prevalence of major BSI pathogens, prominent mechanisms of BSI pathogenesis, opportunities for prevention and diagnosis, and treatment options.
Collapse
Affiliation(s)
- Caitlyn L Holmes
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Owen R Albin
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Harry L T Mobley
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael A Bachman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
3
|
Jahan TA, Lapin NA, O'Connell MT, Jo C, Ma Y, Tareen NG, Copley LA. Accelerated Severity of Illness Score Enhances Prediction of Complicated Acute Hematogenous Osteomyelitis in Children. Pediatr Infect Dis J 2024:00006454-990000000-01001. [PMID: 39259854 DOI: 10.1097/inf.0000000000004535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
BACKGROUND Severity of illness determination for children with acute hematogenous osteomyelitis should be accomplished during the earliest stages of evaluation to guide treatment and establish prognosis. This study objectively defines an outcome of complicated osteomyelitis and explores an illness severity-based model with an improved ability to predict this outcome as soon and accurately as possible, comparing it to existing models. METHODS Children with Staphylococcus aureus acute hematogenous osteomyelitis (n = 438) were retrospectively studied to identify adverse events and predictors of severity. The outcome of complicated osteomyelitis was ultimately defined as the occurrence of any major or at least 3 minor adverse events, which occurred in 52 children. Twenty-four clinical and laboratory predictors were evaluated through univariate and stacked multivariable regression analyses of chronologically distinct groups of variables. Receiver operating characteristic curve analyses were conducted to compare models. RESULTS Accelerated Severity of Illness Score included: triage tachycardia [odds ratio: 10.2 (95% confidence interval: 3.48-32.3], triage tachypnea [6.0 (2.4-15.2)], C-reactive proteininitial ≥17.2 mg/dL [4.5 (1.8-11.8)], white blood cell count band percentageinitial >3.8% [4.6 (2.0-11.0)], hemoglobininitial ≤10.4 g/dL [6.0 (2.6-14.7)], methicillin-resistant S. aureus [3.0 (1.2-8.5)], septic arthritis [4.5 (1.8-12.3)] and platelet nadir [7.2 (2.7-20.4)]. The receiver operating characteristic curve of Accelerated Severity of Illness Score [area under the curve = 0.96 (0.941-0.980)] were superior to those of Modified Severity of Illness Score = 0.903 (0.859-0.947), Acute Score for Complications of Osteomyelitis Risk Evaluation = 0.878 (0.830-0.926) and Chronic Score for Complications of Osteomyelitis Risk Evaluation = 0.858 (0.811-0.904). Successive receiver operating characteristic curve analyses established an exponentially increasing risk of complicated osteomyelitis for children with mild (0/285 or 0%), moderate (4/63 or 6.3%), severe (15/50 or 30.0%) and hyper-severe (33/40 or 82.5%) acute hematogenous osteomyelitis (P<0.0001). CONCLUSIONS This study improves upon previous severity of illness models by identifying early predictors of a rigorously defined outcome of complicated osteomyelitis.
Collapse
Affiliation(s)
- Tahmina A Jahan
- From the Department of Pediatric Infectious Disease, Seattle Children's Hospital, University of Washington, Seattle, Washington
| | - Norman A Lapin
- Department of Orthopaedic Surgery, Texas Scottish Rite Hospital for Children, Center for Pediatric Bone Biology and Translational Research; Dallas, Texas
| | - Michael T O'Connell
- Department of Orthopaedic Surgery, Texas Scottish Rite Hospital for Children, Center for Pediatric Bone Biology and Translational Research; Dallas, Texas
| | - Chanhee Jo
- Department of Clinical Orthopaedic Research, Scottish Rite for Children
| | - Yuhan Ma
- Department of Clinical Orthopaedic Research, Scottish Rite for Children
| | - Naureen G Tareen
- Department of Pediatric Orthopaedic Surgery, Children's Medical Center-Dallas, Dallas, Texas
| | - Lawson A Copley
- Department of Orthopaedic Surgery, Texas Scottish Rite Hospital for Children, Center for Pediatric Bone Biology and Translational Research; Dallas, Texas
- Departments of Orthopaedic Surgery and Pediatrics, University of Texas Southwestern Medical Center
- Department of Pediatric Orthopaedic Surgery, Children's Health System of Texas, Dallas, Texas
| |
Collapse
|
4
|
Tang Y, Zhao J, Suo H, Hu C, Li Q, Li G, Han S, Su X, Song W, Jin M, Li Y, Li S, Wei L, Jiang X, Jiang S. Sinigrin reduces the virulence of Staphylococcus aureus by targeting coagulase. Microb Pathog 2024; 194:106841. [PMID: 39117013 DOI: 10.1016/j.micpath.2024.106841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/26/2024] [Accepted: 08/03/2024] [Indexed: 08/10/2024]
Abstract
Multi-resistant Staphylococcus aureus (S. aureus) infection is a significant global health concern owing to its high mortality and morbidity rates. Coagulase (Coa), a key enzyme that activates prothrombin to initiate host coagulation, has emerged as a promising target for anti-infective therapeutic approaches. This study identified sinigrin as a potent Coa inhibitor that significantly inhibited S. aureus-induced coagulation at concentration as low as 32 mg/L. Additionally, at a higher concentration of 128 mg/L, sinigrin disrupted the self-protection mechanism of S. aureus. Thermal shift and fluorescence-quenching assays confirmed the direct binding of sinigrin to the Coa protein. Molecular docking analysis predicted specific binding sites for sinigrin in the Coa molecule, and point mutation experiments highlighted the importance of Arg-187 and Asp-222 as critical binding sites for both Coa and sinigrin. In vivo studies demonstrated that the combination of sinigrin with oxacillin exhibited greater antibacterial efficacy than oxacillin alone in the treatment of S. aureus-induced pneumonia in mice. Furthermore, sinigrin was shown to reduce bacterial counts and inflammatory cytokine levels in the lung tissues of S. aureus-infected mice. In summary, sinigrin was shown to directly target Coa, resulting in the attenuation of S. aureus virulence, which suggests the potential of sinigrin as an adjuvant for future antimicrobial therapies.
Collapse
Affiliation(s)
- Yating Tang
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Jingming Zhao
- Proctology Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Huiqin Suo
- School of Pharmacy, Changchun University of Traditional Chinese Medicine, Changchun, 130117, China
| | - Chunjie Hu
- Proctology Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Qingjie Li
- PhD Research Center of Traditional Chinese Medicine, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Guofeng Li
- Proctology Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Shaoyu Han
- The University of Queensland, St Lucia, QLD, 4067, China
| | - Xin Su
- School of Basic Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Wu Song
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Mengli Jin
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yufen Li
- School of Basic Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Songyang Li
- School of Pharmacy, Changchun University of Traditional Chinese Medicine, Changchun, 130117, China
| | - Lin Wei
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Xin Jiang
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, 130117, China; School of Basic Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Shuang Jiang
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
5
|
Paul S, Todd OA, Eichelberger KR, Tkaczyk C, Sellman BR, Noverr MC, Cassat JE, Fidel PL, Peters BM. A fungal metabolic regulator underlies infectious synergism during Candida albicans-Staphylococcus aureus intra-abdominal co-infection. Nat Commun 2024; 15:5746. [PMID: 38982056 PMCID: PMC11233573 DOI: 10.1038/s41467-024-50058-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
Candida albicans and Staphylococcus aureus are two commonly associated pathogens that cause nosocomial infections with high morbidity and mortality. Our prior and current work using a murine model of polymicrobial intra-abdominal infection (IAI) demonstrates that synergistic lethality is driven by Candida-induced upregulation of functional S. aureus α-toxin leading to polymicrobial sepsis and organ damage. In order to determine the candidal effector(s) mediating enhanced virulence, an unbiased screen of C. albicans transcription factor mutants was undertaken revealing that zcf13Δ/Δ fails to drive augmented α-toxin or lethal synergism during co-infection. A combination of transcriptional and phenotypic profiling approaches shows that ZCF13 regulates genes involved in pentose metabolism, including RBK1 and HGT7 that contribute to fungal ribose catabolism and uptake, respectively. Subsequent experiments reveal that ribose inhibits the staphylococcal agr quorum sensing system and concomitantly represses toxicity. Unlike wild-type C. albicans, zcf13Δ/Δ did not effectively utilize ribose during co-culture or co-infection leading to exogenous ribose accumulation and agr repression. Forced expression of RBK1 and HGT7 in the zcf13Δ/Δ mutant fully restores pathogenicity during co-infection. Collectively, our results detail the interwoven complexities of cross-kingdom interactions and highlight how intermicrobial metabolism impacts polymicrobial disease pathogenesis with devastating consequences for the host.
Collapse
Affiliation(s)
- Saikat Paul
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Olivia A Todd
- Integrated Program in Biomedical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kara R Eichelberger
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christine Tkaczyk
- Early Vaccines and Immune Therapies, AstraZeneca, Gaithersburg, MD, USA
| | - Bret R Sellman
- Early Vaccines and Immune Therapies, AstraZeneca, Gaithersburg, MD, USA
| | - Mairi C Noverr
- Department of Microbiology and Immunology, School of Medicine, Tulane University, New Orleans, LA, USA
| | - James E Cassat
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paul L Fidel
- Department of Oral and Craniofacial Biology, Louisiana State University Health - School of Dentistry, New Orleans, LA, USA
| | - Brian M Peters
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA.
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
6
|
Mobarakai O, Barrios S, Singh A, Stone J. A rare case of MRSA associated Lemierre's syndrome. IDCases 2024; 37:e02014. [PMID: 39071048 PMCID: PMC11283075 DOI: 10.1016/j.idcr.2024.e02014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024] Open
Abstract
Lemierre's syndrome, or postanginal sepsis, is an uncommon but potentially fatal infection of the internal jugular vein. The combination of bacteremia, internal jugular vein thrombophlebitis, and metastatic septic emboli secondary to acute pharyngeal infections is characteristic of Lemierre's syndrome. Isolated pathogens are typically oral anaerobic bacteria, most commonly Fusobacterium necrophorum. While the incidence of Lemierre's syndrome has declined over the years, the proportion of cases caused by uncommonly implicated bacteria have been increasingly cited in the literature (1). In this case report, we introduce a novel presentation of Lemierre's syndrome in a patient who presented to the emergency department with neck swelling and shortness of breath and was found to have infectious myositis and bacteremia with methicillin-resistant Staphylococcus aureus. Clinicians should be vigilant of underlying thrombus in patients with neck swelling and infectious myositis as our patient's internal jugular vein thrombus was missed on initial computed tomography read.
Collapse
Affiliation(s)
- Olivia Mobarakai
- Pennsylvania Hospital of the University of Pennsylvania Health System, United States
| | - Sofia Barrios
- Pennsylvania Hospital of the University of Pennsylvania Health System, United States
| | - Ajit Singh
- Pennsylvania Hospital of the University of Pennsylvania Health System, United States
| | - Jarle Stone
- Pennsylvania Hospital of the University of Pennsylvania Health System, United States
| |
Collapse
|
7
|
Barber MF, Fitzgerald JR. Mechanisms of host adaptation by bacterial pathogens. FEMS Microbiol Rev 2024; 48:fuae019. [PMID: 39003250 PMCID: PMC11308195 DOI: 10.1093/femsre/fuae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/02/2024] [Accepted: 07/24/2024] [Indexed: 07/15/2024] Open
Abstract
The emergence of new infectious diseases poses a major threat to humans, animals, and broader ecosystems. Defining factors that govern the ability of pathogens to adapt to new host species is therefore a crucial research imperative. Pathogenic bacteria are of particular concern, given dwindling treatment options amid the continued expansion of antimicrobial resistance. In this review, we summarize recent advancements in the understanding of bacterial host species adaptation, with an emphasis on pathogens of humans and related mammals. We focus particularly on molecular mechanisms underlying key steps of bacterial host adaptation including colonization, nutrient acquisition, and immune evasion, as well as suggest key areas for future investigation. By developing a greater understanding of the mechanisms of host adaptation in pathogenic bacteria, we may uncover new strategies to target these microbes for the treatment and prevention of infectious diseases in humans, animals, and the broader environment.
Collapse
Affiliation(s)
- Matthew F Barber
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, United States
- Department of Biology, University of Oregon, Eugene, OR 97403, United States
| | - J Ross Fitzgerald
- The Roslin Institute, University of Edinburgh, Midlothian, EH25 9RG, United Kingdom
| |
Collapse
|
8
|
Yu Y, Yang M, Zhao H, Zhang C, Liu K, Liu J, Li C, Cai B, Guan F, Yao M. Natural blackcurrant extract contained gelatin hydrogel with photothermal and antioxidant properties for infected burn wound healing. Mater Today Bio 2024; 26:101113. [PMID: 38933414 PMCID: PMC11201118 DOI: 10.1016/j.mtbio.2024.101113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Burns represent a prevalent global health concern and are particularly susceptible to bacterial infections. Severe infections may lead to serious complications, posing a life-threatening risk. Near-infrared (NIR)-assisted photothermal antibacterial combined with antioxidant hydrogel has shown significant potential in the healing of infected wounds. However, existing photothermal agents are typically metal-based, complicated to synthesize, or pose biosafety hazards. In this study, we utilized plant-derived blackcurrant extract (B) as a natural source for both photothermal and antioxidant properties. By incorporating B into a G-O hydrogel crosslinked through Schiff base reaction between gelatin (G) and oxidized pullulan (O), the resulting G-O-B hydrogel exhibited good injectability and biocompatibility along with robust photothermal and antioxidant activities. Upon NIR irradiation, the controlled temperature (around 45-50 °C) generated by the G-O-B hydrogel resulted in rapid (10 min) and efficient killing of Staphylococcus aureus (99 %), Escherichia coli (98 %), and Pseudomonas aeruginosa (82 %). Furthermore, the G-O-B0.5 hydrogel containing 0.5 % blackcurrant extract promoted collagen deposition, angiogenesis, and accelerated burn wound closure conclusively, demonstrating that this well-designed and extract-contained hydrogel dressing holds immense potential for enhancing the healing process of bacterial-infected burn wounds.
Collapse
Affiliation(s)
- Yachao Yu
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, China
| | - Mengyu Yang
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, China
| | - Hua Zhao
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, China
| | - Chen Zhang
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, China
| | - Kaiyue Liu
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, China
| | - Jingmei Liu
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, China
| | - Chenghao Li
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, China
| | - Bingjie Cai
- Department of Dermatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fangxia Guan
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, China
| | - Minghao Yao
- School of Life Science, Zhengzhou University, 100 Science Road, Zhengzhou, 450001, China
| |
Collapse
|
9
|
Subsomwong P, Teng W, Ishiai T, Narita K, Sukchawalit R, Nakane A, Asano K. Extracellular vesicles from Staphylococcus aureus promote the pathogenicity of Pseudomonas aeruginosa. Microbiol Res 2024; 281:127612. [PMID: 38244256 DOI: 10.1016/j.micres.2024.127612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/22/2024]
Abstract
Co-infections with Staphylococcus aureus and Pseudomonas aeruginosa are common in patients with chronic wounds, but little is known about their synergistic effect mediated by extracellular vesicles (EVs). In this study, we investigated the effect of EVs derived from S. aureus (SaEVs) on the pathogenicity of P. aeruginosa. By using lipophilic dye, we could confirm the fusion between SaEV and P. aeruginosa membranes. However, SaEVs did not alter the growth and antibiotic susceptible pattern of P. aeruginosa. Differential proteomic analysis between SaEV-treated and non-treated P. aeruginosa was performed, and the results revealed that lipopolysaccharide (LPS) biosynthesis protein in P. aeruginosa significantly increased after SaEV-treatment. Regarding this result, we also found that SaEVs promoted LPS production, biofilm formation, and expression of polysaccharide polymerization-related genes in P. aeruginosa. Furthermore, invasion of epithelial cells by SaEV-pretreated P. aeruginosa was enhanced. On the other hand, uptake of P. aeruginosa by RAW 264.7 macrophages was impaired after pretreatment P. aeruginosa with SaEVs. Proteomic analysis SaEVs revealed that SaEVs contain the proteins involving in host cell colonization, inhibition of host immune response, anti-phagocytosis of the macrophages, and protein translocation and iron uptake of S. aureus. In conclusion, SaEVs serve as a mediator that promote P. aeruginosa pathogenicity by enhancing LPS biosynthesis, biofilm formation, epithelial cell invasion, and macrophage uptake impairment.
Collapse
Affiliation(s)
- Phawinee Subsomwong
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Wei Teng
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Takahito Ishiai
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kouji Narita
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan; Institute for Animal Experimentation, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Rojana Sukchawalit
- Laboratory of Biotechnology, Chulabhorn Research Institute, Lak Si, Bangkok, Thailand
| | - Akio Nakane
- Department of Biopolymer and Health Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Krisana Asano
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan; Department of Biopolymer and Health Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan.
| |
Collapse
|
10
|
Che D, Hu J, Zhu J, Lyu J, Zhang X. Development and validation of a nomogram for predicting in-hospital mortality in ICU patients with infective endocarditis. BMC Med Inform Decis Mak 2024; 24:84. [PMID: 38515185 PMCID: PMC10958908 DOI: 10.1186/s12911-024-02482-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 03/11/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Infective endocarditis (IE) is a disease with high in-hospital mortality. The objective of the present investigation was to develop and validate a nomogram that precisely anticipates in-hospital mortality in ICU individuals diagnosed with infective endocarditis. METHODS Retrospectively collected clinical data of patients with IE admitted to the ICU in the MIMIC IV database were analyzed using the Least Absolute Shrinkage and Selection Operator (LASSO) regression to identify potential hazards. A logistic regression model incorporating multiple factors was established, and a dynamic nomogram was generated to facilitate predictions. To assess the classification performance of the model, an ROC curve was generated, and the AUC value was computed as an indicator of its diagnostic accuracy. The model was subjected to calibration curve analysis and the Hosmer-Lemeshow (HL) test to assess its goodness of fit. To evaluate the clinical relevance of the model, decision-curve analysis (DCA) was conducted. RESULTS The research involved a total of 676 patients, who were divided into two cohorts: a training cohort comprising 473 patients and a validation cohort comprising 203 patients. The allocation ratio between the two cohorts was 7:3. Based on the independent predictors identified through LASSO regression, the final selection for constructing the prediction model included five variables: lactate, bicarbonate, white blood cell count (WBC), platelet count, and prothrombin time (PT). The nomogram model demonstrated a robust diagnostic ability in both the cohorts used for training and validation. This is supported by the respective area under the curve (AUC) values of 0.843 and 0.891. The results of the calibration curves and HL tests exhibited acceptable conformity between observed and predicted outcomes. According to the DCA analysis, the nomogram model demonstrated a notable overall clinical advantage compared to the APSIII and SAPSII scoring systems. CONCLUSIONS The nomogram developed during the study proved to be highly accurate in forecasting the mortality of patients with IE during hospitalization in the ICU. As a result, it may be useful for clinicians in decision-making and treatment.
Collapse
Affiliation(s)
- Dongyang Che
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Jinan University, 510630, Guangzhou, Guangdong Province, People's Republic of China
| | - Jinlin Hu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Guangzhou, Guangdong Provincial Hospital of Chinese Medicine, University of Chinese Medicine, 510630, Guangzhou, Guangdong Province, People's Republic of China
| | - Jialiang Zhu
- The First Affiliated Hospital of Jinan University, 510630, Guangzhou, Guangdong Province, People's Republic of China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, 510630, Guangzhou, Guangdong Province, People's Republic of China.
| | - Xiaoshen Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Jinan University, 510630, Guangzhou, Guangdong Province, People's Republic of China.
| |
Collapse
|
11
|
Paul S, Todd OA, Eichelberger KR, Tkaczyk C, Sellman BR, Noverr MC, Cassat JE, Fidel PL, Peters BM. A fungal metabolic regulator underlies infectious synergism during Candida albicans - Staphylococcus aureus intra-abdominal co-infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.15.580531. [PMID: 38405692 PMCID: PMC10888754 DOI: 10.1101/2024.02.15.580531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Candida albicans and Staphylococcus aureus are two commonly associated pathogens that cause nosocomial infections with high morbidity and mortality. Our prior and current work using a murine model of polymicrobial intra-abdominal infection (IAI) uncovered synergistic lethality that was driven by Candida -induced upregulation of functional S. aureus ⍺-toxin leading to polymicrobial sepsis and organ damage. In order to determine the candidal effector(s) mediating enhanced virulence, an unbiased screen of C. albicans transcription factor mutants was undertaken and revealed that zcf13 Δ/Δ failed to drive augmented ⍺-toxin or lethal synergism during co-infection. Using a combination of transcriptional and phenotypic profiling approaches, ZCF13 was shown to regulate genes involved in pentose metabolism, including RBK1 and HGT7 that contribute to fungal ribose catabolism and uptake, respectively. Subsequent experiments revealed that ribose inhibited the staphylococcal agr quorum sensing system and concomitantly repressed toxicity. Unlike wild-type C. albicans , zcf13 Δ/Δ was unable to effectively utilize ribose during co-culture or co-infection leading to exogenous ribose accumulation and agr repression. Forced expression of RBK1 and HGT7 in the zcf13 Δ/Δ mutant fully restored pathogenicity during co-infection. Collectively, our results detail the interwoven complexities of cross-kingdom interactions and highlight how intermicrobial metabolism impacts polymicrobial disease pathogenesis with devastating consequences for the host.
Collapse
|
12
|
Edwards JV, Prevost NT, Hinchliffe DJ, Nam S, Chang S, Hron RJ, Madison CA, Smith JN, Poffenberger CN, Taylor MM, Martin EJ, Dixon KJ. Preparation and Activity of Hemostatic and Antibacterial Dressings with Greige Cotton/Zeolite Formularies Having Silver and Ascorbic Acid Finishes. Int J Mol Sci 2023; 24:17115. [PMID: 38069435 PMCID: PMC10706952 DOI: 10.3390/ijms242317115] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/01/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
The need for prehospital hemostatic dressings that exert an antibacterial effect is of interest for prolonged field care. Here, we consider a series of antibacterial and zeolite formulary treatment approaches applied to a cotton-based dressing. The design of the fabric formulations was based on the hemostatic dressing TACGauze with zeolite Y incorporated as a procoagulant with calcium and pectin to facilitate fiber adherence utilizing silver nanoparticles, and cellulose-crosslinked ascorbic acid to confer antibacterial activity. Infra-red spectra were employed to characterize the chemical modifications on the dressings. Contact angle measurements were employed to document the surface hydrophobicity of the cotton fabric which plays a role in the contact activation of the coagulation cascade. Ammonium Y zeolite-treated dressings initiated fibrin equal to the accepted standard hemorrhage control dressing and showed similar improvement with antibacterial finishes. The antibacterial activity of cotton-based technology utilizing both citrate-linked ascorbate-cellulose conjugate analogs and silver nanoparticle-embedded cotton fibers was observed against Staphylococcus aureus and Klebsiella pneumoniae at a level of 99.99 percent in the AATCC 100 assay. The hydrogen peroxide levels of the ascorbic acid-based fabrics, measured over a time period from zero up to forty-eight hours, were in line with the antibacterial activities.
Collapse
Affiliation(s)
- J. Vincent Edwards
- Southern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, New Orleans, LA 70124, USA; (N.T.P.); (D.J.H.); (S.N.); (S.C.); (R.J.H.); (C.A.M.); (J.N.S.)
| | - Nicolette T. Prevost
- Southern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, New Orleans, LA 70124, USA; (N.T.P.); (D.J.H.); (S.N.); (S.C.); (R.J.H.); (C.A.M.); (J.N.S.)
| | - Doug J. Hinchliffe
- Southern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, New Orleans, LA 70124, USA; (N.T.P.); (D.J.H.); (S.N.); (S.C.); (R.J.H.); (C.A.M.); (J.N.S.)
| | - Sunghyun Nam
- Southern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, New Orleans, LA 70124, USA; (N.T.P.); (D.J.H.); (S.N.); (S.C.); (R.J.H.); (C.A.M.); (J.N.S.)
| | - SeChin Chang
- Southern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, New Orleans, LA 70124, USA; (N.T.P.); (D.J.H.); (S.N.); (S.C.); (R.J.H.); (C.A.M.); (J.N.S.)
| | - Rebecca J. Hron
- Southern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, New Orleans, LA 70124, USA; (N.T.P.); (D.J.H.); (S.N.); (S.C.); (R.J.H.); (C.A.M.); (J.N.S.)
| | - Crista A. Madison
- Southern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, New Orleans, LA 70124, USA; (N.T.P.); (D.J.H.); (S.N.); (S.C.); (R.J.H.); (C.A.M.); (J.N.S.)
| | - Jade N. Smith
- Southern Regional Research Center, Agricultural Research Service, United States Department of Agriculture, New Orleans, LA 70124, USA; (N.T.P.); (D.J.H.); (S.N.); (S.C.); (R.J.H.); (C.A.M.); (J.N.S.)
| | - Chelsie N. Poffenberger
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (C.N.P.); (M.M.T.); (K.J.D.)
| | - Michelle M. Taylor
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (C.N.P.); (M.M.T.); (K.J.D.)
| | - Erika J. Martin
- Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Kirsty J. Dixon
- Department of Surgery, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA; (C.N.P.); (M.M.T.); (K.J.D.)
| |
Collapse
|
13
|
Pezzanite LM, Chow L, Dow SW, Goodrich LR, Gilbertie JM, Schnabel LV. Antimicrobial Properties of Equine Stromal Cells and Platelets and Future Directions. Vet Clin North Am Equine Pract 2023; 39:565-578. [PMID: 37442729 DOI: 10.1016/j.cveq.2023.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2023] Open
Abstract
Increasing antimicrobial resistance in veterinary practice has driven the investigation of novel therapeutic strategies including regenerative and biologic therapies to treat bacterial infection. Integration of biological approaches such as platelet lysate and mesenchymal stromal cell (MSC) therapy may represent adjunctive treatment strategies for bacterial infections that minimize systemic side effects and local tissue toxicity associated with traditional antibiotics and that are not subject to antibiotic resistance. In this review, we will discuss mechanisms by which biological therapies exert antimicrobial effects, as well as potential applications and challenges in clinical implementation in equine practice.
Collapse
Affiliation(s)
- Lynn M Pezzanite
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| | - Lyndah Chow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Steven W Dow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA; Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Laurie R Goodrich
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Jessica M Gilbertie
- Department of Microbiology and Immunology, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA
| | - Lauren V Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, North Carolina State University, Raleigh, NC, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
14
|
Martens CP, Peetermans M, Vanassche T, Verhamme P, Jacquemin M, Martinod K. Peptidylarginine deiminase 4 and ADAMTS13 activity in Staphylococcus aureus bacteraemia. Philos Trans R Soc Lond B Biol Sci 2023; 378:20230042. [PMID: 37778390 PMCID: PMC10542450 DOI: 10.1098/rstb.2023.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/05/2023] [Indexed: 10/03/2023] Open
Abstract
Staphylococcus aureus infection is associated with increased levels of neutrophil extracellular traps (NETs) and von Willebrand factor (VWF), and with reduced activity of ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motifs, member 13). Peptidylarginine deiminase 4 (PAD4) contributes to NET formation and inactivates ADAMTS13 in vitro. The role of PADs in the dynamics of NETs, VWF and ADAMTS13 has not yet been studied. We thus aimed to assess the longitudinal evolution of NETs, PADs, VWF and ADAMTS13 activity in S. aureus infection. Plasma samples from S. aureus bacteraemia patients were longitudinally collected and analysed for NETs, PAD4/PAD2, VWF and ADAMTS13 activity. Correlation analyses with clinical data were performed. Recombinant PAD4 and S. aureus were assessed in vitro for their potential to modulate ADAMTS13 activity. Sixty-seven patients were included. Plasma levels of NETs, VWF, PAD4 and PAD2 were increased and ADAMTS13 activity was decreased. Levels of PADs were negatively correlated with ADAMTS13 activity. NETs were positively correlated with PADs, and negatively with ADAMTS13 activity. In vitro, recombinant PAD4 but not S. aureus reduced ADAMTS13 activity in plasma. Levels of PAD4 and PAD2 correlate with reduced ADAMTS13 activity, with neutrophils as the likely source of PAD activity in S. aureus bacteraemia. This article is part of the Theo Murphy meeting issue 'The virtues and vices of protein citrullination'.
Collapse
Affiliation(s)
- Caroline P. Martens
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Immunology and Transplantation, KU Leuven, Leuven, 3000, Belgium
| | - Marijke Peetermans
- Laboratory for Clinical Infectious and Inflammatory Diseases, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, 3000, Belgium
- Medical Intensive Care Unit, Department of General Internal Medicine, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Thomas Vanassche
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Immunology and Transplantation, KU Leuven, Leuven, 3000, Belgium
- Department of Cardiovascular Diseases, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Peter Verhamme
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Immunology and Transplantation, KU Leuven, Leuven, 3000, Belgium
- Department of Cardiovascular Diseases, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Marc Jacquemin
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Immunology and Transplantation, KU Leuven, Leuven, 3000, Belgium
| | - Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, Immunology and Transplantation, KU Leuven, Leuven, 3000, Belgium
| |
Collapse
|
15
|
Hou J, Jiang Y, Xu Y, Zhao C, Cao Y, Song W, Wang B. Inhibitory effect of bilobalide on Staphylococcus aureus von Willebrand factor-binding protein and its therapeutic effect in mice with pneumonia. J Appl Microbiol 2023; 134:lxad233. [PMID: 37833234 DOI: 10.1093/jambio/lxad233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/13/2023] [Accepted: 10/12/2023] [Indexed: 10/15/2023]
Abstract
AIMS Disabling bacterial virulence with small molecules has been proposed as a potential strategy to prevent bacterial pathogenicity. The von Willebrand factor-binding protein of Staphylococcus aureus was identified previously as a key virulence determinant. Our objective was to discover a von Willebrand-factor binding protein (vWbp) inhibitor distinct from the antibiotics used to prevent infections resulting from S. aureus. METHODS AND RESULTS Using coagulation assays, we found that the sesquiterpene trilactone bilobalide blocks coagulation mediated by vWbp, but has no impact on the growth of S. aureus at a concentration of 128 μg ml-1. Moreover, a mouse model of pneumonia caused by S. aureus indicated that bilobalide could attenuate S. aureus virulence in vivo. This effect is achieved not by interfering with the expression of vWbp but by binding to vWbp, as demonstrated by western blotting, thermal shift assays, and fluorescence quenching assays. Using molecular dynamic simulations and point mutagenesis analysis, we identified that the Q17A and R453A residues are key residues for the binding of bilobalide to vWbp. CONCLUSIONS Overall, we tested the ability of bilobalide to inhibit S. aureus infections by targeting vWbp and explored the potential mechanism of this activity.
Collapse
Affiliation(s)
- Juan Hou
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Yijing Jiang
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Yangming Xu
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Chunhui Zhao
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Yali Cao
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
- Changchun University of Traditional Chinese Medicine Second Affiliated Hospital, Changchun University of Chinese Medicine, Changchun 130052, China
| | - Wu Song
- School of Clinical Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| | - Bingmei Wang
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130017, China
| |
Collapse
|
16
|
Parente R, Fumagalli MR, Di Claudio A, Cárdenas Rincón CL, Erreni M, Zanini D, Iapichino G, Protti A, Garlanda C, Rusconi R, Doni A. A Multilayered Imaging and Microfluidics Approach for Evaluating the Effect of Fibrinolysis in Staphylococcus aureus Biofilm Formation. Pathogens 2023; 12:1141. [PMID: 37764949 PMCID: PMC10534389 DOI: 10.3390/pathogens12091141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
The recognition of microbe and extracellular matrix (ECM) is a recurring theme in the humoral innate immune system. Fluid-phase molecules of innate immunity share regulatory roles in ECM. On the other hand, ECM elements have immunological functions. Innate immunity is evolutionary and functionally connected to hemostasis. Staphylococcus aureus (S. aureus) is a major cause of hospital-associated bloodstream infections and the most common cause of several life-threatening conditions such as endocarditis and sepsis through its ability to manipulate hemostasis. Biofilm-related infection and sepsis represent a medical need due to the lack of treatments and the high resistance to antibiotics. We designed a method combining imaging and microfluidics to dissect the role of elements of the ECM and hemostasis in triggering S. aureus biofilm by highlighting an essential role of fibrinogen (FG) in adhesion and formation. Furthermore, we ascertained an important role of the fluid-phase activation of fibrinolysis in inhibiting biofilm of S. aureus and facilitating an antibody-mediated response aimed at pathogen killing. The results define FG as an essential element of hemostasis in the S. aureus biofilm formation and a role of fibrinolysis in its inhibition, while promoting an antibody-mediated response. Understanding host molecular mechanisms influencing biofilm formation and degradation is instrumental for the development of new combined therapeutic approaches to prevent the risk of S. aureus biofilm-associated diseases.
Collapse
Affiliation(s)
- Raffaella Parente
- Multiscale ImmunoImaging Unit (mIIu), IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Maria Rita Fumagalli
- Multiscale ImmunoImaging Unit (mIIu), IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Alessia Di Claudio
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Italy
| | - Cindy Lorena Cárdenas Rincón
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Italy
| | - Marco Erreni
- Multiscale ImmunoImaging Unit (mIIu), IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Italy
| | - Damiano Zanini
- Multiscale ImmunoImaging Unit (mIIu), IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Giacomo Iapichino
- Department of Anesthesia and Intensive Care Units, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Alessandro Protti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Italy
- Department of Anesthesia and Intensive Care Units, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Cecilia Garlanda
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Roberto Rusconi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Andrea Doni
- Multiscale ImmunoImaging Unit (mIIu), IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| |
Collapse
|
17
|
Ferreira RM, Dos Santos Silva DH, Silva KF, de Melo Monteiro J, Ferreira GF, Silva MRC, da Silva LCN, de Castro Oliveira L, Monteiro AS. Draft genome sequence of Staphylococcus aureus sequence type 5 SA01 isolated from bloodstream infection and comparative analysis with reference strains. Funct Integr Genomics 2023; 23:288. [PMID: 37653266 DOI: 10.1007/s10142-023-01204-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023]
Abstract
A Staphylococcus aureus isolate (SA01) obtained from bloodstream infection exhibited a remarkable drug resistance profile. In this study, we report the draft genome sequence of S. aureus ST 5 SA01, a multidrug-resistant isolate, and analyzed the genes associated with drug resistance and virulence. The genome sketch of S. aureus ST5 SA01 was sequenced with Illumina and annotated using the Prokka software. Rapid Annotation Subsystem Technology (RAST) was used to verify the gene functions in the genome subsystems. The Comprehensive Antibiotic Resistance Database (CARD) and Virulence Factor Database (VFDB) were used in the analysis. The RAST indicated a contribution of 25 proteins to host adenine, fibronectin-binding protein A (FnbA), and biofilm formation as an intercellular polysaccharide adhesive system (PIA). The MLST indicated that S. aureus ST 5 SA01 belongs to ST5 (CC5). In silico analyses also showed an extensive repertoire of genes associated with toxins, such as LukGH leukocidin, enterotoxins, and superantigen staphylococcal classes (SSL). The 11 genes for antimicrobial resistance in S. aureus ST 5 SA01 showed similarity and identity above ≥ 99% with nucleotide sequences deposited in GenBank. Although studies on ST5 clones in Brazil are scarce, monitoring the clone of S. aureus ST 5 SA01 is essential, as it has become a problem in pediatrics in several countries.
Collapse
Affiliation(s)
- Romulo Maia Ferreira
- Laboratório de Microbiologia Aplicada, Universidade CEUMA, São Luís, 65075-120, MA, Brasil
| | | | - Karinny Farias Silva
- Laboratório de Microbiologia Aplicada, Universidade CEUMA, São Luís, 65075-120, MA, Brasil
| | | | - Gabriella Freitas Ferreira
- Departamento de Farmácia, Universidade Federal de Juiz de Fora - Campus Governador Valadares, CEP 35010-180, Juiz de Fora, MG, Brasil
| | | | | | - Letícia de Castro Oliveira
- Departamento de Microbiologia, Universidade Federal Do Triângulo Mineiro, Imunologia E Parasitologia, 38025180, Uberaba, MG, Brasil
| | - Andrea Souza Monteiro
- Laboratório de Microbiologia Aplicada, Universidade CEUMA, São Luís, 65075-120, MA, Brasil
| |
Collapse
|
18
|
Lu X, Yang M, Zhou S, Yang S, Chen X, Khalid M, Wang K, Fang Y, Wang C, Lai R, Duan Z. Identification and Characterization of RK22, a Novel Antimicrobial Peptide from Hirudinaria manillensis against Methicillin Resistant Staphylococcus aureus. Int J Mol Sci 2023; 24:13453. [PMID: 37686259 PMCID: PMC10487658 DOI: 10.3390/ijms241713453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Staphylococcus aureus (S. aureus) infections are a leading cause of morbidity and mortality, which are compounded by drug resistance. By manipulating the coagulation system, S. aureus gains a significant advantage over host defense mechanisms, with hypercoagulation induced by S. aureus potentially aggravating infectious diseases. Recently, we and other researchers identified that a higher level of LL-37, one endogenous antimicrobial peptide with a significant killing effect on S. aureus infection, resulted in thrombosis formation through the induction of platelet activation and potentiation of the coagulation factor enzymatic activity. In the current study, we identified a novel antimicrobial peptide (RK22) from the salivary gland transcriptome of Hirudinaria manillensis (H. manillensis) through bioinformatic analysis, and then synthesized it, which exhibited good antimicrobial activity against S. aureus, including a clinically resistant strain with a minimal inhibitory concentration (MIC) of 6.25 μg/mL. The RK22 peptide rapidly killed S. aureus by inhibiting biofilm formation and promoting biofilm eradication, with good plasma stability, negligible cytotoxicity, minimal hemolytic activity, and no significant promotion of the coagulation system. Notably, administration of RK22 significantly inhibited S. aureus infection and the clinically resistant strain in vivo. Thus, these findings highlight the potential of RK22 as an ideal treatment candidate against S. aureus infection.
Collapse
Affiliation(s)
- Xiaoyu Lu
- School of Life Sciences, Tianjin University, Tianjin 300072, China;
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
| | - Min Yang
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Shengwen Zhou
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Shuo Yang
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xiran Chen
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Mehwish Khalid
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Kexin Wang
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- School of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Yaqun Fang
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
| | - Chaoming Wang
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Ren Lai
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650107, China
- National Resource for Non-Human Primates, Kunming Primate Research Center, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650107, China
- Sino-African Joint Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Zilei Duan
- Key Laboratory of Bioactive Peptides of Yunnan Province, National & Local Joint Engineering Center of Natural Bioactive Peptides, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; (M.Y.); (S.Z.); (S.Y.); (X.C.); (M.K.); (K.W.); (Y.F.); (C.W.)
| |
Collapse
|
19
|
Unar A, Bertolino L, Patauner F, Gallo R, Durante-Mangoni E. Pathophysiology of Disseminated Intravascular Coagulation in Sepsis: A Clinically Focused Overview. Cells 2023; 12:2120. [PMID: 37681852 PMCID: PMC10486945 DOI: 10.3390/cells12172120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/07/2023] [Accepted: 08/12/2023] [Indexed: 09/09/2023] Open
Abstract
Sepsis is a major global health problem that results from a dysregulated and uncontrolled host response to infection, causing organ failure. Despite effective anti-infective therapy and supportive treatments, the mortality rate of sepsis remains high. Approximately 30-80% of patients with sepsis may develop disseminated intravascular coagulation (DIC), which can double the mortality rate. There is currently no definitive treatment approach for sepsis, with etiologic treatment being the cornerstone of therapy for sepsis-associated DIC. Early detection, diagnosis, and treatment are critical factors that impact the prognosis of sepsis-related DIC. Over the past several decades, researchers have made continuous efforts to better understand the mechanisms of DIC in sepsis, as well as improve its quantitative diagnosis and treatment. This article aims to provide a comprehensive overview of the current understanding of sepsis-related DIC, focusing on common causes and diagnoses, with the goal of guiding healthcare providers in the care of patients with sepsis.
Collapse
Affiliation(s)
- Ahsanullah Unar
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, 80138 Naples, Italy; (A.U.); (L.B.); (F.P.); (R.G.)
| | - Lorenzo Bertolino
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, 80138 Naples, Italy; (A.U.); (L.B.); (F.P.); (R.G.)
| | - Fabian Patauner
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, 80138 Naples, Italy; (A.U.); (L.B.); (F.P.); (R.G.)
| | - Raffaella Gallo
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, 80138 Naples, Italy; (A.U.); (L.B.); (F.P.); (R.G.)
| | - Emanuele Durante-Mangoni
- Department of Precision Medicine, University of Campania ‘L. Vanvitelli’, 80138 Naples, Italy; (A.U.); (L.B.); (F.P.); (R.G.)
- Unit of Infectious and Transplant Medicine, AORN Ospedali dei Colli-Monaldi Hospital, 80131 Naples, Italy
| |
Collapse
|
20
|
Komorowicz E, Farkas VJ, Szabó L, Cherrington S, Thelwell C, Kolev K. DNA and histones impair the mechanical stability and lytic susceptibility of fibrin formed by staphylocoagulase. Front Immunol 2023; 14:1233128. [PMID: 37662916 PMCID: PMC10470048 DOI: 10.3389/fimmu.2023.1233128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Background Staphylocoagulase (SCG) is a virulence factor of Staphylococcus aureus, one of the most lethal pathogens of our times. The complex of SCG with prothrombin (SCG/ProT) can clot fibrinogen, and SCG/ProT-induced fibrin and plasma clots have been described to show decreased mechanical and lytic resistance, which may contribute to septic emboli from infected cardiac vegetations. At infection sites, neutrophils can release DNA and histones, as parts of neutrophil extracellular traps (NETs), which in turn favor thrombosis, inhibit fibrinolysis and strengthen clot structure. Objectives To characterize the combined effects of major NET-components (DNA, histone H1 and H3) on SCG/ProT-induced clot structure, mechanical and lytic stability. Methods Recombinant SCG was used to clot purified fibrinogen and plasma. The kinetics of formation and lysis of fibrin and plasma clots containing H1 or core histones+/-DNA were followed by turbidimetry. Fibrin structure and mechanical stability were characterized with scanning electron microscopy, pressure-driven permeation, and oscillation rheometry. Results Histones and DNA favored the formation of thicker fibrin fibers and a more heterogeneous clot structure including high porosity with H1 histone, whereas low porosity with core histones and DNA. As opposed to previous observations with thrombin-induced clots, SCG/ProT-induced fibrin was not mechanically stabilized by histones. Similarly to thrombin-induced clots, the DNA-histone complexes prolonged fibrinolysis with tissue-type plasminogen activator (up to 2-fold). The anti-fibrinolytic effect of the DNA and DNA-H3 complex was observed in plasma clots too. Heparin (low molecular weight) accelerated the lysis of SCG/ProT-clots from plasma, even if DNA and histones were also present. Conclusions In the interplay of NETs and fibrin formed by SCG, DNA and histones promote structural heterogeneity in the clots, and fail to stabilize them against mechanical stress. The DNA-histone complexes render the SCG-fibrin more resistant to lysis and thereby less prone to embolization.
Collapse
Affiliation(s)
- Erzsébet Komorowicz
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - Veronika J. Farkas
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - László Szabó
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
- Plasma Chemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Budapest, Hungary
| | - Sophie Cherrington
- South Mimms Laboratories, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | - Craig Thelwell
- South Mimms Laboratories, Medicines and Healthcare Products Regulatory Agency, Potters Bar, United Kingdom
| | - Krasimir Kolev
- Institute of Biochemistry and Molecular Biology, Department of Biochemistry, Semmelweis University, Budapest, Hungary
| |
Collapse
|
21
|
Ariëns RAS, Cassat JE. Surviving a sticky situation: therapeutic administration of fibrinogen variant γ' improves outcomes of Staphylococcus aureus septicemia. J Thromb Haemost 2023; 21:2048-2050. [PMID: 37468174 PMCID: PMC10947783 DOI: 10.1016/j.jtha.2023.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 07/21/2023]
Affiliation(s)
- Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - James E Cassat
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
22
|
Vago JP, Zaidan I, Perucci LO, Brito LF, Teixeira LC, Silva CMS, Miranda TC, Melo EM, Bruno AS, Queiroz-Junior CM, Sugimoto MA, Tavares LP, Grossi LC, Borges IN, Schneider AH, Baik N, Schneider AH, Talvani A, Ferreira RG, Alves-Filho JC, Nobre V, Teixeira MM, Parmer RJ, Miles LA, Sousa LP. Plasmin and plasminogen prevent sepsis severity by reducing neutrophil extracellular traps and systemic inflammation. JCI Insight 2023; 8:e166044. [PMID: 36917195 PMCID: PMC10243804 DOI: 10.1172/jci.insight.166044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
Sepsis is a lethal syndrome characterized by systemic inflammation and abnormal coagulation. Despite therapeutic advances, sepsis mortality remains substantially high. Herein, we investigated the role of the plasminogen/plasmin (Plg/Pla) system during sepsis. Plasma levels of Plg were significantly lower in mice subjected to severe compared with nonsevere sepsis, whereas systemic levels of IL-6, a marker of sepsis severity, were higher in severe sepsis. Plg levels correlated negatively with IL-6 in both septic mice and patients, whereas plasminogen activator inhibitor-1 levels correlated positively with IL-6. Plg deficiency render mice susceptible to nonsevere sepsis induced by cecal ligation and puncture (CLP), resulting in greater numbers of neutrophils and M1 macrophages, liver fibrin(ogen) deposition, lower efferocytosis, and increased IL-6 and neutrophil extracellular trap (NET) release associated with organ damage. Conversely, inflammatory features, fibrin(ogen), and organ damage were substantially reduced, and efferocytosis was increased by exogenous Pla given during CLP- and LPS-induced endotoxemia. Plg or Pla protected mice from sepsis-induced lethality and enhanced the protective effect of antibiotics. Mechanistically, Plg/Pla-afforded protection was associated with regulation of NET release, requiring Pla-protease activity and lysine binding sites. Plg/Pla are important host-protective players during sepsis, controlling local and systemic inflammation and collateral organ damage.
Collapse
Affiliation(s)
- Juliana P. Vago
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Isabella Zaidan
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| | - Luiza O. Perucci
- Department of Biological Sciences, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Larissa Froede Brito
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| | - Lívia C.R. Teixeira
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| | - Camila Meirelles Souza Silva
- Department of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Thaís C. Miranda
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| | - Eliza M. Melo
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alexandre S. Bruno
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michelle A. Sugimoto
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana P. Tavares
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laís C. Grossi
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| | - Isabela N. Borges
- Hospital of Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ayda Henriques Schneider
- Department of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Nagyung Baik
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Ayda H. Schneider
- Department of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - André Talvani
- Department of Biological Sciences, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Raphael G. Ferreira
- Department of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - José C. Alves-Filho
- Department of Pharmacology, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Vandack Nobre
- Hospital of Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M. Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Robert J. Parmer
- Department of Medicine, Veterans Administration San Diego Healthcare System and University of California, San Diego, California, USA
| | - Lindsey A. Miles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Lirlândia P. Sousa
- Signaling in Inflammation Laboratory, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, and
| |
Collapse
|
23
|
Meyers S, Lox M, Kraisin S, Liesenborghs L, Martens CP, Frederix L, Van Bruggen S, Crescente M, Missiakas D, Baatsen P, Vanassche T, Verhamme P, Martinod K. Neutrophils Protect Against Staphylococcus aureus Endocarditis Progression Independent of Extracellular Trap Release. Arterioscler Thromb Vasc Biol 2023; 43:267-285. [PMID: 36453281 PMCID: PMC9869964 DOI: 10.1161/atvbaha.122.317800] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Infective endocarditis (IE) is characterized by an infected thrombus at the heart valves. How bacteria bypass the immune system and cause these thrombi remains unclear. Neutrophils releasing NETs (neutrophil extracellular traps) lie at this interface between host defense and coagulation. We aimed to determine the role of NETs in IE immunothrombosis. METHODS We used a murine model of Staphylococcus aureus endocarditis in which IE is provoked on inflamed heart valves and characterized IE thrombus content by immunostaining identifying NETs. Antibody-mediated neutrophil depletion and neutrophil-selective PAD4 (peptidylarginine deiminase 4)-knockout mice were used to clarify the role of neutrophils and NETs, respectively. S. aureus mutants deficient in key virulence factors related to immunothrombosis (nucleases or staphylocoagulases) were investigated. RESULTS Neutrophils releasing NETs were present in infected thrombi and within cellular infiltrates in the surrounding vasculature. Neutrophil depletion increased occurrence of IE, whereas neutrophil-selective impairment of NET formation did not alter IE occurrence. Absence of S. aureus nuclease, which degrades NETs, did not affect endocarditis outcome. In contrast, absence of staphylocoagulases (coagulase and von Willebrand factor binding protein) led to improved survival, decreased bacteremia, smaller infiltrates, and decreased tissue destruction. Significantly more NETs were present in these vegetations, which correlated with decreased bacteria and cell death in the adjacent vascular wall. CONCLUSIONS Neutrophils protect against IE independent of NET release. Absence of S. aureus coagulases, but not nucleases, reduced IE severity and increased NET levels. Staphylocoagulase-induced fibrin likely hampers NETs from constraining infection and the resultant tissue damage, a hallmark of valve destruction in IE.
Collapse
Affiliation(s)
- Severien Meyers
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences (S.M., M.L., S.K., L.L., C.P.M., L.F., S.V.B., T.V., P.V., K.M.), KU Leuven, Belgium
| | - Marleen Lox
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences (S.M., M.L., S.K., L.L., C.P.M., L.F., S.V.B., T.V., P.V., K.M.), KU Leuven, Belgium
| | - Sirima Kraisin
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences (S.M., M.L., S.K., L.L., C.P.M., L.F., S.V.B., T.V., P.V., K.M.), KU Leuven, Belgium
| | - Laurens Liesenborghs
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences (S.M., M.L., S.K., L.L., C.P.M., L.F., S.V.B., T.V., P.V., K.M.), KU Leuven, Belgium
| | - Caroline P. Martens
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences (S.M., M.L., S.K., L.L., C.P.M., L.F., S.V.B., T.V., P.V., K.M.), KU Leuven, Belgium
| | - Liesbeth Frederix
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences (S.M., M.L., S.K., L.L., C.P.M., L.F., S.V.B., T.V., P.V., K.M.), KU Leuven, Belgium
| | - Stijn Van Bruggen
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences (S.M., M.L., S.K., L.L., C.P.M., L.F., S.V.B., T.V., P.V., K.M.), KU Leuven, Belgium
| | - Marilena Crescente
- Department of Life Sciences, Manchester Metropolitan University, United Kingdom (M.C.)
| | | | - Pieter Baatsen
- Electron Microscopy-Platform of the VIB Bio Imaging Core and VIB Center for Brain and Disease Research (P.B.), KU Leuven, Belgium
| | - Thomas Vanassche
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences (S.M., M.L., S.K., L.L., C.P.M., L.F., S.V.B., T.V., P.V., K.M.), KU Leuven, Belgium
| | - Peter Verhamme
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences (S.M., M.L., S.K., L.L., C.P.M., L.F., S.V.B., T.V., P.V., K.M.), KU Leuven, Belgium
| | - Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences (S.M., M.L., S.K., L.L., C.P.M., L.F., S.V.B., T.V., P.V., K.M.), KU Leuven, Belgium
| |
Collapse
|
24
|
Van Bruggen S, Martinod K. The coming of age of neutrophil extracellular traps in thrombosis: Where are we now and where are we headed? Immunol Rev 2022; 314:376-398. [PMID: 36560865 DOI: 10.1111/imr.13179] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thrombosis remains a major problem in our society, manifesting across multiple demographic groups and with high associated morbidity and mortality. Thrombus development is the result of a complex mechanism in which multiple cell types and soluble factors play a crucial role. One cell that has gained the most attention in recent years is the neutrophil. This key member of the innate immune system can form neutrophil extracellular traps (NETs) in response to activating stimuli in circulation. NETs form a scaffold for thrombus formation, both initiating the process and stabilizing the final product. As the first responders of the host immune system, neutrophils have the flexibility to recognize a variety of molecules and can quickly interact with a range of different cell types. This trait makes them sensitive to exogenous stimuli. NET formation in response to pathogens is well established, leading to immune-mediated thrombus formation or immunothrombosis. NETs can also be formed during sterile inflammation through the activation of neutrophils by fellow immune cells including platelets, or activated endothelium. In chronic inflammatory settings, NETs can ultimately promote the development of tissue fibrosis, with organ failure as an end-stage outcome. In this review, we discuss the different pathways through which neutrophils can be activated toward NET formation and how these processes can result in a shared outcome: thrombus formation. Finally, we evaluate these different interactions and mechanisms for their potential as therapeutic targets, with neutrophil-targeted therapies providing a future approach to treating thrombosis. In contrast to current practices, such treatment could result in reduced pathogenic blood clot formation without increasing the risk of bleeding.
Collapse
Affiliation(s)
- Stijn Van Bruggen
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Risser F, López-Morales J, Nash MA. Adhesive Virulence Factors of Staphylococcus aureus Resist Digestion by Coagulation Proteases Thrombin and Plasmin. ACS BIO & MED CHEM AU 2022; 2:586-599. [PMID: 36573096 PMCID: PMC9782320 DOI: 10.1021/acsbiomedchemau.2c00042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 12/30/2022]
Abstract
Staphylococcus aureus (S. aureus) is an invasive and life-threatening pathogen that has undergone extensive coevolution with its mammalian hosts. Its molecular adaptations include elaborate mechanisms for immune escape and hijacking of the coagulation and fibrinolytic pathways. These capabilities are enacted by virulence factors including microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) and the plasminogen-activating enzyme staphylokinase (SAK). Despite the ability of S. aureus to modulate coagulation, until now the sensitivity of S. aureus virulence factors to digestion by proteases of the coagulation system was unknown. Here, we used protein engineering, biophysical assays, and mass spectrometry to study the susceptibility of S. aureus MSCRAMMs to proteolytic digestion by human thrombin, plasmin, and plasmin/SAK complexes. We found that MSCRAMMs were highly resistant to proteolysis, and that SAK binding to plasmin enhanced this resistance. We mapped thrombin, plasmin, and plasmin/SAK cleavage sites of nine MSCRAMMs and performed biophysical, bioinformatic, and stability analysis to understand structural and sequence features common to protease-susceptible sites. Overall, our study offers comprehensive digestion patterns of S. aureus MSCRAMMs by thrombin, plasmin, and plasmin/SAK complexes and paves the way for new studies into this resistance and virulence mechanism.
Collapse
Affiliation(s)
- Fanny Risser
- Institute
of Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland,Department
of Biosystems Sciences and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Joanan López-Morales
- Institute
of Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland,Department
of Biosystems Sciences and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Michael A. Nash
- Institute
of Physical Chemistry, Department of Chemistry, University of Basel, 4058 Basel, Switzerland,Department
of Biosystems Sciences and Engineering, ETH Zurich, 4058 Basel, Switzerland,E-mail:
| |
Collapse
|
26
|
Abstract
OBJECTIVES We aimed to describe the variation of hemostasis proteins in children with bacterial infections due to different pathogens ( Neisseria meningitidis, Streptococcus pneumoniae, Staphylococcus aureus , and group A streptococcus [GAS]) and to study hemostasis proteins in relation to mortality. DESIGN Preplanned analysis in prospective cohort study. SETTING Hospitals in five European countries (Austria, The Netherlands, Spain, Switzerland, and the United Kingdom). PATIENTS Admitted children (2012-2016) with community-acquired infections due to meningococci ( n = 83), pneumococci ( n = 64), S. aureus (n = 50), and GAS ( n = 44) with available serum samples collected less than 48 hours after admission. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Fibronectin, plasminogen activator inhibitor type 1 (PAI-1), thrombomodulin, and a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS-13) were measured in serum in 2019-2020. Additionally, von Willebrand factor, protein C, protein S, and factor IX were measured in citrate plasma available from a subset of patients. Outcome measures included in-hospital mortality and disease severity (need for ventilation/inotropes, Pediatric Index of Mortality score).Of 241 children, 21 (8.7%) died and 177 (73.5%) were admitted to PICU. Mortality rate was similar for the pathogen groups. Levels of fibronectin and thrombomodulin differed for the different pathogens ( p < 0.05). Fibronectin levels were lower in GAS infections than in S. pneumoniae and S. aureus infections but did not differ from meningococcal infections. Thrombomodulin levels in meningococcal infections were higher than in S. aureus and pneumococcal infections. Overall, the area under the curve for mortality was 0.81 (95% CI, 0.70-0.92) for thrombomodulin and 0.78 (95% CI, 0.69-0.88) for ADAMTS-13. The association of each hemostasis protein did not vary across pathogens for any of the outcome measures. CONCLUSIONS Hemostatic disturbances in childhood bacterial infections are not limited to meningococcal sepsis but occur with a comparable severity across nonmeningococcal infections. High thrombomodulin and high ADAMTS-13 had good discriminative ability for mortality. Our results emphasize the importance of hemostatic disturbances in meningococcal and nonmeningococcal pediatric bacterial infections.
Collapse
|
27
|
Frias-De-Diego A, Gilbertie JM, Scholle F, Dejarnette S, Crisci E. Effect of BIO-PLY TM, a Platelet-Rich Plasma Derived Biologic on PRRSV-2-Infected Macrophages. Viruses 2022; 14:v14122666. [PMID: 36560670 PMCID: PMC9783555 DOI: 10.3390/v14122666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022] Open
Abstract
Porcine Reproductive and Respiratory Syndrome (PRRS) is the one of the most devastating diseases impacting the swine industry worldwide. Control and prevention methods rely on biosafety measures and vaccination. As an RNA virus with a high rate of mutation, vaccines are only partially effective against circulating and newly emerging strains. To reduce the burden of this disease, research on alternative control methods is needed. Here, we assess the in vitro antiviral effect of a novel platelet-rich plasma-derived biologic termed BIO-PLYTM (for the BIOactive fraction of Platelet-rich plasma LYsate) from both swine and equine origin. Our results show that BIO-PLYTM significantly reduces the amount of PRRSV viral load determined by RT-qPCR and the number of infectious viral particles measured by TCID50 in infected porcine alveolar and parenchymal macrophages. This study also showed limited toxicity of BIO-PLYTM in vitro and aspects of its immunomodulatory capacity evaluating the regulation of reactive oxygen species and cytokines production in infected cells. Finally, this study presents promising data on the effect of BIO-PLYTM on other RNA viruses such as human A influenza viruses and coronavirus.
Collapse
Affiliation(s)
- Alba Frias-De-Diego
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Jessica M. Gilbertie
- Department of Biomedical Affairs and Research, Edward Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA
| | - Frank Scholle
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Sarah Dejarnette
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Elisa Crisci
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
- Correspondence: ; Tel.: +1-919-513-6255
| |
Collapse
|
28
|
Zheng M, Zhu K, Peng H, Shang W, Zhao Y, Lu S, Rao X, Li M, Zhou R, Li G. CcpA Regulates Staphylococcus aureus Biofilm Formation through Direct Repression of Staphylokinase Expression. Antibiotics (Basel) 2022; 11:antibiotics11101426. [PMID: 36290085 PMCID: PMC9598941 DOI: 10.3390/antibiotics11101426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Staphylococcus aureus represents a notorious opportunistic pathogen causing various infections in biofilm nature, imposing remarkable therapeutic challenges worldwide. The catabolite control protein A (CcpA), a major regulator of carbon catabolite repression (CCR), has been recognized to modulate S. aureus biofilm formation, while the underlying mechanism remains to be fully elucidated. In this study, the reduced biofilm was firstly determined in the ccpA deletion mutant of S. aureus clinical isolate XN108 using both crystal violet staining and confocal laser scanning microscopy. RNA-seq analysis suggested that sak-encoding staphylokinase (Sak) was significantly upregulated in the mutant ∆ccpA, which was further confirmed by RT-qPCR. Consistently, the induced Sak production correlated the elevated promoter activity of sak and increased secretion in the supernatants, as demonstrated by Psak-lacZ reporter fusion expression and chromogenic detection, respectively. Notably, electrophoretic mobility shift assays showed that purified recombinant protein CcpA binds directly to the promoter region of sak, suggesting the direct negative control of sak expression by CcpA. Double isogenic deletion of ccpA and sak restored biofilm formation for mutant ∆ccpA, which could be diminished by trans-complemented sak. Furthermore, the exogenous addition of recombinant Sak inhibited biofilm formation for XN108 in a dose-dependent manner. Together, this study delineates a novel model of CcpA-controlled S. aureus biofilm through direct inhibition of sak expression, highlighting the multifaceted roles and multiple networks regulated by CcpA.
Collapse
Affiliation(s)
- Mingxia Zheng
- Department of Emergency Medicine, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Keting Zhu
- Department of Emergency Medicine, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Huagang Peng
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Weilong Shang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Yan Zhao
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Shuguang Lu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
| | - Ming Li
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
- Correspondence: (M.L.); (R.Z.); (G.L.)
| | - Renjie Zhou
- Department of Emergency Medicine, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
- Correspondence: (M.L.); (R.Z.); (G.L.)
| | - Gang Li
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Chongqing 400038, China
- Correspondence: (M.L.); (R.Z.); (G.L.)
| |
Collapse
|
29
|
Comparative Genotypic Analysis of RAPD and RFLP Markers for Molecular Variation Detection of Methicillin-Resistant Staphylococcus aureus Clinical Isolates. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58091245. [PMID: 36143922 PMCID: PMC9505472 DOI: 10.3390/medicina58091245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/25/2022]
Abstract
Background and Objectives: Methicillin-resistant Staphylococcus aureus (MRSA) isolates are associated with various diseases ranged from mild superficial impairments to invasive infections. This study aimed to evaluate the ability of polymerase chain reaction (PCR) based methods namely, restriction fragment length polymorphism (RFLP) of the coa gene and random amplified polymorphic DNA (RAPD), to determine the genetic diversity of MRSA isolates. Materials and Methods: A total of 37 MRSA isolates were conventionally identified depending on their biochemical and microbiological culture characteristics. Genotypic confirmation was based on detection of the associated mecA gene. The genetic variation amongst MRSA isolates was evaluated following the coa gene-based RFLP and RAPD fingerprints. Results: Results illustrated that, the species specific coa gene was detected in all MRSA isolates. The irregular bands intensity, number, and molecular sizes of the PCR amplicons demonstrated the coa gene polymorphism. The incompatible AluI digestion patterns of these amplicons classified the tested MRSA isolates into 20 RFLP patterns which confirm the coa gene polymorphism. Additionally, the PCR-based RAPD analysis showed variable bands number with size range of approximately 130 bp to 4 kbp, which indicated the genetic variation of the tested MRSA isolates as it created 36 variable RAPD banding profiles. Conclusions: coa gene AluI enzymatic restriction sites, amongst the tested MRSA isolates, certify their genetic variation on the basis of the accurate but complicated and relatively expensive coa gene-based RFLP. Conversely, the results verified the excellent ability of the simple and cost-effective PCR-based RAPD analysis to discriminate between MRSA isolates without any preface data about the genome.
Collapse
|
30
|
Staphylococcus aureus increases platelet reactivity in patients with infective endocarditis. Sci Rep 2022; 12:12933. [PMID: 35902612 PMCID: PMC9334290 DOI: 10.1038/s41598-022-16681-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/13/2022] [Indexed: 01/30/2023] Open
Abstract
Thromboembolism is frequent in infective endocarditis (IE). However, the optimal antithrombotic regimen in IE is unknown. Staphylococcus aureus (SA) is the leading cause of IE. First studies emphasize increased platelet reactivity by SA. In this pilot study, we hypothesized that platelet reactivity is increased in patients with SA− IE, which could be abrogated by antiplatelet medication. We conducted a prospective, observatory, single-center cohort study in 114 patients with IE, with four cohorts: (1) SA coagulase positive IE without aspirin (ASA) medication, (2) coagulase negative IE without ASA, (3) SA coagulase positive IE with ASA, (4) coagulase negative IE with ASA. Platelet function was measured by Multiplate electrode aggregometry, blood clotting by ROTEM thromboelastometry. Bleeding events were assessed according to TIMI classification. In ASA-naïve patients, aggregation with ADP was increased with coag. pos. IE (coagulase negative: 39.47 ± 4.13 AUC vs. coagulase positive: 59.46 ± 8.19 AUC, p = 0.0219). This was abrogated with ASA medication (coagulase negative: 42.4 ± 4.67 AUC vs. coagulase positive: 45.11 ± 6.063 AUC p = 0.7824). Aspirin did not increase bleeding in SA positive patients. However, in SA negative patients with aspirin, red blood cell transfusions were enhanced. SA coagulase positive IE is associated with increased platelet reactivity. This could be abrogated by aspirin without increased bleeding risk. The results of this pilot study suggest that ASA might be beneficial in SA coagulase positive IE. This needs to be confirmed in clinical trials.
Collapse
|
31
|
Pisaryuk AS, Zamarashkina VA, Safarova NB, Povalyaev NM, Kotova EO, Babukhina UI, Koltsova EM, Kobalava ZD. Coagulation Disorders in Infective Endocarditis: Role of Pathogens, Biomarkers, Antithrombotic Therapy (Systematic Review). RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2022. [DOI: 10.20996/1819-6446-2022-06-14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The issue of antithrombotic therapy in patients with infective endocarditis has been studied for over 75 years. During that time studying of pathogenesis of the disease and its embolic complications, lead to the introduction of the concept of “immunothrombosis”. That mechanism allows infective agents (mostly bacteria) to be cloaked from the immune system and to multiply freely, leading to growth of vegetation, thus resulting in higher chance of fragmentation. Small-scale experimental and clinical studies on the correction of hemostatic disorders in infective endocarditis, that were performed in 20th century, didn’t show any significant results, that could affect clinical practice. However, reinterpretation of available data on coagulative system will allow to have elements of hemostasis as an application point in treating infective endocarditis. The article will discuss latest insights on the role of hemostasis system in pathophysisology of infective endocarditis, its effects on the development of the embolic complications, perspectives for diagnostics and treatment.
Collapse
Affiliation(s)
- A. S. Pisaryuk
- RUDN University; Moscow City Hospital named after V.V. Vinogradov
| | | | | | | | | | | | - E. M. Koltsova
- Center for Theoretical Problems of Physicochemical Pharmacology; Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology
| | | |
Collapse
|
32
|
Berry KA, Verhoef MTA, Leonard AC, Cox G. Staphylococcus aureus adhesion to the host. Ann N Y Acad Sci 2022; 1515:75-96. [PMID: 35705378 DOI: 10.1111/nyas.14807] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Staphylococcus aureus is a pathobiont capable of colonizing and infecting most tissues within the human body, resulting in a multitude of different clinical outcomes. Adhesion of S. aureus to the host is crucial for both host colonization and the establishment of infections. Underlying the pathogen's success is a complex and diverse arsenal of adhesins. In this review, we discuss the different classes of adhesins, including a consideration of the various adhesion sites throughout the body and the clinical outcomes of each infection type. The development of therapeutics targeting the S. aureus host-pathogen interaction is a relatively understudied area. Due to the increasing global threat of antimicrobial resistance, it is crucial that innovative and alternative approaches are considered. Neutralizing virulence factors, through the development of antivirulence agents, could reduce bacterial pathogenicity and the ever-increasing burden of S. aureus infections. This review provides insight into potentially efficacious adhesion-associated targets for the development of novel decolonizing and antivirulence strategies.
Collapse
Affiliation(s)
- Kirsten A Berry
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Mackenzie T A Verhoef
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Allison C Leonard
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Georgina Cox
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
33
|
Oberbach A, Schlichting N, Hagl C, Lehmann S, Kullnick Y, Friedrich M, Köhl U, Horn F, Kumbhari V, Löffler B, Schmidt F, Joskowiak D, Born F, Saha S, Bagaev E. Four decades of experience of prosthetic valve endocarditis reflect a high variety of diverse pathogens. Cardiovasc Res 2022; 119:410-428. [PMID: 35420122 DOI: 10.1093/cvr/cvac055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/04/2022] [Accepted: 03/23/2022] [Indexed: 01/18/2023] Open
Abstract
Prosthetic valve endocarditis (PVE) remains a serious condition with a high mortality rate. Precise identification of the PVE-associated pathogen/s and their virulence is essential for successful therapy, and patient survival. The commonly described PVE-associated pathogens are staphylococci, streptococci and enterococci, with Staphylococcus aureus being the most frequently diagnosed species. Furthermore, multi-drug resistance pathogens are increasing in prevalence, and continue to pose new challenges mandating a personalized approach. Blood cultures in combination with echocardiography are the most common methods to diagnose PVE, often being the only indication, it exists. In many cases, the diagnostic strategy recommended in the clinical guidelines does not identify the precise microbial agent and to frequently, false negative blood cultures are reported. Despite the fact that blood culture findings are not always a good indicator of the actual PVE agent in the valve tissue, only a minority of re-operated prostheses are subjected to microbiological diagnostic evaluation. In this review, we focus on the diversity and the complete spectrum of PVE-associated bacterial, fungal and viral pathogens in blood, and prosthetic heart valve, their possible virulence potential, and their challenges in making a microbial diagnosis. We are curious to understand if the unacceptable high mortality of PVE is associated with the high number of negative microbial findings in connection with a possible PVE. Herein, we discuss the possibilities and limits of the diagnostic methods conventionally used and make recommendations for enhanced pathogen identification. We also show possible virulence factors of the most common PVE-associated pathogens and their clinical effects. Based on blood culture, molecular biological diagnostics, and specific valve examination, better derivations for the antibiotic therapy as well as possible preventive intervention can be established in the future.
Collapse
Affiliation(s)
- Andreas Oberbach
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany.,Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Nadine Schlichting
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Christian Hagl
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany.,Munich Heart Alliance, Partner Site German Centre for Cardiovascular Disease (DZHK), Munich, Germany
| | - Stefanie Lehmann
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Yvonne Kullnick
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Maik Friedrich
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Ulrike Köhl
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Friedemann Horn
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Vivek Kumbhari
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Florida, USA
| | - Bettina Löffler
- Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Frank Schmidt
- Proteomics Core, Weill Cornell Medical Centre Qatar, Doha, Qatar
| | - Dominik Joskowiak
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany
| | - Frank Born
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany
| | - Shekhar Saha
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany
| | - Erik Bagaev
- Department of Cardiac Surgery, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
34
|
Engineered Molecular Therapeutics Targeting Fibrin and the Coagulation System: a Biophysical Perspective. Biophys Rev 2022; 14:427-461. [PMID: 35399372 PMCID: PMC8984085 DOI: 10.1007/s12551-022-00950-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023] Open
Abstract
The coagulation cascade represents a sophisticated and highly choreographed series of molecular events taking place in the blood with important clinical implications. One key player in coagulation is fibrinogen, a highly abundant soluble blood protein that is processed by thrombin proteases at wound sites, triggering self-assembly of an insoluble protein hydrogel known as a fibrin clot. By forming the key protein component of blood clots, fibrin acts as a structural biomaterial with biophysical properties well suited to its role inhibiting fluid flow and maintaining hemostasis. Based on its clinical importance, fibrin is being investigated as a potentially valuable molecular target in the development of coagulation therapies. In this topical review, we summarize our current understanding of the coagulation cascade from a molecular, structural and biophysical perspective. We highlight single-molecule studies on proteins involved in blood coagulation and report on the current state of the art in directed evolution and molecular engineering of fibrin-targeted proteins and polymers for modulating coagulation. This biophysical overview will help acclimatize newcomers to the field and catalyze interdisciplinary work in biomolecular engineering toward the development of new therapies targeting fibrin and the coagulation system.
Collapse
|
35
|
Meyers S, Crescente M, Verhamme P, Martinod K. Staphylococcus aureus and Neutrophil Extracellular Traps: The Master Manipulator Meets Its Match in Immunothrombosis. Arterioscler Thromb Vasc Biol 2022; 42:261-276. [PMID: 35109674 PMCID: PMC8860219 DOI: 10.1161/atvbaha.121.316930] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the past 10 years, neutrophil extracellular traps (NETs) have become widely accepted as an integral player in immunothrombosis, due to their complex interplay with both pathogens and components of the coagulation system. While the release of NETs is an attempt by neutrophils to trap pathogens and constrain infections, NETs can have bystander effects on the host by inducing uncontrolled thrombosis, inflammation, and tissue damage. From an evolutionary perspective, pathogens have adapted to bypass the host innate immune response. Staphylococcus aureus (S. aureus), in particular, proficiently overcomes NET formation using several virulence factors. Here we review mechanisms of NET formation and how these are intertwined with platelet activation, the release of endothelial von Willebrand factor, and the activation of the coagulation system. We discuss the unique ability of S. aureus to modulate NET formation and alter released NETs, which helps S. aureus to escape from the host's defense mechanisms. We then discuss how platelets and the coagulation system could play a role in NET formation in S. aureus-induced infective endocarditis, and we explain how targeting these complex cellular interactions could reveal novel therapies to treat this disease and other immunothrombotic disorders.
Collapse
Affiliation(s)
- Severien Meyers
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Belgium (S.M., M.C., P.V., K.M.)
| | - Marilena Crescente
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Belgium (S.M., M.C., P.V., K.M.).,Department of Life Sciences, Manchester Metropolitan University, United Kingdom (M.C.)
| | - Peter Verhamme
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Belgium (S.M., M.C., P.V., K.M.)
| | - Kimberly Martinod
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Belgium (S.M., M.C., P.V., K.M.)
| |
Collapse
|
36
|
Feng R, Dan N, Chen Y, Dan W. Crosslinking of dialdehyde heparin: a new strategy for improving the anticoagulant properties of porcine acellular dermal matrix. RSC Adv 2022; 12:6811-6820. [PMID: 35424614 PMCID: PMC8981558 DOI: 10.1039/d1ra08982j] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/22/2022] [Indexed: 01/25/2023] Open
Abstract
The anticoagulant properties of valve materials are essential to maintain blood patency after artificial valve implantation. Porcine acellular dermal matrix (pADM) has low immunogenicity, good biocompatibility, and can reduce calcification by eliminating heterogeneous cells. However, its main component is collagen, which has strong coagulation function and poor anticoagulant activity. When used in heart valve materials, it can easily coagulate and form a life-threatening thrombus. Therefore, it is necessary to improve its anticoagulant performance. The glutaraldehyde (GA) cross-linked valves widely used clinically are easy to calcify with poor anticoagulant performance and cytotoxicity. In this study, dialdehyde heparin containing cross-linking active aldehyde groups was prepared by sodium periodate oxidation, then it was used for crosslinking with pADM to chemically modify its anticoagulant performance. Compared with GA cross-linked pADM (GA-pA), dialdehyde heparin cross-linked pADM (OL-pA) has better thermal stability and biocompatibility, especially its anticoagulant and antiplatelet adhesion were significantly improved, which can reduce the incidence of coagulation, thrombocytopenia and bleeding. In summary, dialdehyde heparin is expected to be applied to modify the anticoagulant properties of pADM and has great potential for the preparation and clinical application of anticoagulant materials such as heart valves and artificial blood vessels.
Collapse
Affiliation(s)
- Rongxin Feng
- Key Laboratory of Leather Chemistry and Engineering of the Education Ministry, Sichuan University Chengdu Sichuan 610065 China
| | - Nianhua Dan
- Key Laboratory of Leather Chemistry and Engineering of the Education Ministry, Sichuan University Chengdu Sichuan 610065 China
- Research Center of Biomedical Engineering, Sichuan University Chengdu Sichuan 610065 China
| | - Yining Chen
- Key Laboratory of Leather Chemistry and Engineering of the Education Ministry, Sichuan University Chengdu Sichuan 610065 China
- Research Center of Biomedical Engineering, Sichuan University Chengdu Sichuan 610065 China
| | - Weihua Dan
- Key Laboratory of Leather Chemistry and Engineering of the Education Ministry, Sichuan University Chengdu Sichuan 610065 China
- Research Center of Biomedical Engineering, Sichuan University Chengdu Sichuan 610065 China
| |
Collapse
|
37
|
Cuervo G, Hernández-Meneses M, Falces C, Quintana E, Vidal B, Marco F, Perissinotti A, Carratalà J, Miro JM. Infective Endocarditis: New Challenges in a Classic Disease. Semin Respir Crit Care Med 2022; 43:150-172. [PMID: 35172365 DOI: 10.1055/s-0042-1742482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Infective endocarditis is a relatively rare, but deadly infection, with an overall mortality of around 20% in most series. Clinical manifestations have evolved in response to significant epidemiological shifts in industrialized nations, with a move toward a nosocomial or health-care-related pattern, in older patients, with more episodes associated with prostheses and/or intravascular electronic devices and a predominance of staphylococcal and enterococcal etiology.Diagnosis is often challenging and is based on the conjunction of clinical, microbiological, and imaging information, with notable progress in recent years in the accuracy of echocardiographic data, coupled with the recent emergence of other useful imaging techniques such as cardiac computed tomography (CT) and nuclear medicine tools, particularly 18F-fluorodeoxyglucose positron emission/CT.The choice of an appropriate treatment for each specific case is complex, both in terms of the selection of the appropriate agent and doses and durations of therapy as well as the possibility of using combined bactericidal antibiotic regimens in the initial phase and finalizing treatment at home in patients with good evolution with outpatient oral or parenteral antimicrobial therapies programs. A relevant proportion of patients will also require valve surgery during the active phase of treatment, the timing of which is extremely difficult to define. For all the above, the management of infective endocarditis requires a close collaboration of multidisciplinary endocarditis teams.
Collapse
Affiliation(s)
- Guillermo Cuervo
- Infectious Diseases Service, Hospital Bellvitge - IDIBELL, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Hernández-Meneses
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.,Infectious Diseases Service, Hospital Clinic - IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Carles Falces
- Cardiology Service, Hospital Clinic - IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Eduard Quintana
- Cardiovascular Surgery Service, Hospital Clinic - IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Bárbara Vidal
- Cardiology Service, Hospital Clinic - IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Francesc Marco
- Microbiology Service, Hospital Clinic - IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Andrés Perissinotti
- Department of Nuclear Medicine, Hospital Clinic - IDIBAPS, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Jordi Carratalà
- Infectious Diseases Service, Hospital Bellvitge - IDIBELL, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jose M Miro
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.,Infectious Diseases Service, Hospital Clinic - IDIBAPS, University of Barcelona, Barcelona, Spain
| | | |
Collapse
|
38
|
Abstract
The management of infective endocarditis is complex and inherently requires multidisciplinary cooperation. About half of all patients diagnosed with infective endocarditis will meet the criteria to undergo cardiac surgery, which regularly takes place in urgent or emergency settings. The pathophysiology and clinical presentation of infective endocarditis make it a unique disorder within cardiac surgery that warrants a thorough understanding of specific characteristics in the perioperative period. This includes, among others, echocardiography, coagulation, bleeding management, or treatment of organ dysfunction. In this narrative review article, the authors summarize the current knowledge on infective endocarditis relevant for the clinical anesthesiologist in perioperative management of respective patients. Furthermore, the authors advocate for the anesthesiologist to become a structural member of the endocarditis team.
Collapse
|
39
|
Fusaroli M, Raschi E, Gatti M, De Ponti F, Poluzzi E. Development of a Network-Based Signal Detection Tool: The COVID-19 Adversome in the FDA Adverse Event Reporting System. Front Pharmacol 2021; 12:740707. [PMID: 34955821 PMCID: PMC8694570 DOI: 10.3389/fphar.2021.740707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/18/2021] [Indexed: 12/24/2022] Open
Abstract
Introduction: The analysis of pharmacovigilance databases is crucial for the safety profiling of new and repurposed drugs, especially in the COVID-19 era. Traditional pharmacovigilance analyses-based on disproportionality approaches-cannot usually account for the complexity of spontaneous reports often with multiple concomitant drugs and events. We propose a network-based approach on co-reported events to help assessing disproportionalities and to effectively and timely identify disease-, comorbidity- and drug-related syndromes, especially in a rapidly changing low-resources environment such as that of COVID-19. Materials and Methods: Reports on medications administered for COVID-19 were extracted from the FDA Adverse Event Reporting System quarterly data (January-September 2020) and queried for disproportionalities (Reporting Odds Ratio corrected for multiple comparisons). A network (the Adversome) was estimated considering events as nodes and conditional co-reporting as links. Communities of significantly co-reported events were identified. All data and scripts employed are available in a public repository. Results: Among the 7,082 COVID-19 reports extracted, the seven most frequently suspected drugs (remdesivir, hydroxychloroquine, azithromycin, tocilizumab, lopinavir/ritonavir, sarilumab, and ethanol) have shown disproportionalities with 54 events. Of interest, myasthenia gravis with hydroxychloroquine, and cerebrovascular vein thrombosis with azithromycin. Automatic clustering identified 13 communities, including a methanol-related neurotoxicity associated with alcohol-based hand-sanitizers and a long QT/hepatotoxicity cluster associated with azithromycin, hydroxychloroquine and lopinavir-ritonavir interactions. Conclusion: Findings from the Adversome detect plausible new signals and iatrogenic syndromes. Our network approach complements traditional pharmacovigilance analyses, and may represent a more effective signal detection technique to guide clinical recommendations by regulators and specific follow-up confirmatory studies.
Collapse
Affiliation(s)
| | | | | | | | - Elisabetta Poluzzi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
40
|
Theys H, Van den Eynde J, Herregods MC, Moreillon P, Heying R, Oosterlinck W. The role of antiaggregant agents and anticoagulants in the prevention of aortic valve endocarditis: A double-cohort retrospective study. JTCVS OPEN 2021; 8:301-312. [PMID: 36004157 PMCID: PMC9390520 DOI: 10.1016/j.xjon.2021.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 10/19/2021] [Indexed: 11/23/2022]
Abstract
Objective Antiaggregants (Ag) could prevent infective endocarditis (IE) in preclinical studies. In this study we investigated whether Ag or anticoagulants (Ac) were also protective in humans. Methods In part I we determined the incidence of IE of bioprosthetic aortic valves (PVE) in 333 consecutive patients who underwent aortic valve replacement for noninfective aortic insufficiency between 2009 and 2019. In part II we retrospectively analyzed data of 137 patients who had developed IE of the native aortic valve (NVE) between 2007 and 2015. Multivariable Fine–Gray and logistic regression models were used to investigate associations between Ag and Ac therapy and IE. Results Sixteen of 333 (4.8%) aortic valve replacement recipients developed PVE after a median of 3.72 years. There was no association between Ag and PVE, whereas Ac was associated with a higher IE occurrence (no association for vitamin K antagonists but significant for fondaparinux or low molecular-weight heparins; hazard ratio, 4.61; 95% CI, 1.01-21.9). In contrast, among the 137 patients in part II, vitamin K antagonists (odds ratio [OR], 7.52; 95% CI, 2.51-22.6), double antiplatelet therapy (OR, 44.3; 95% CI, 4.83-407), novel oral Ac (OR, 4.17; 95% CI, 1.15-15.1), and fondaparinux or low molecular-weight heparins (OR, 9.87; 95% CI, 1.81-53.9), but not acetylsalicylic acid, were associated with NVE. Conclusions Ac were associated with IE in both cohorts, whereas Ag were not associated with PVE. This might reflect differences in the studied populations, with Ag and Ac being prescribed for conditions associated with long-term IE risk in the NVE cohort. Therefore, determining the potential protective effect of Ag and Ac will necessitate further well–controlled studies.
Collapse
Affiliation(s)
- Hanne Theys
- Department of Cardiovascular Diseases, Research Unit of Cardiac Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Jef Van den Eynde
- Department of Cardiovascular Diseases, Research Unit of Cardiac Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Helen B. Taussig Heart Center, The Johns Hopkins Hospital and School of Medicine, Baltimore, Md
| | - Marie-Christine Herregods
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Philippe Moreillon
- Department of Fundamental Microbiology, University Lausanne, Lausanne, Switzerland
| | - Ruth Heying
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Department of Pediatric and Congenital Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Wouter Oosterlinck
- Department of Cardiovascular Diseases, Research Unit of Cardiac Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Address for reprints: Wouter Oosterlinck, MD, PhD, Department of Cardiovascular Diseases, University Hospitals Leuven, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
41
|
Dunkel B. Hypercoagulation and thrombosis associated with infection in horses. EQUINE VET EDUC 2021. [DOI: 10.1111/eve.13598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- B. Dunkel
- Department of Veterinary Clinical Sciences The Royal Veterinary College Equine Referral Hospital Hatfield Hertfordshire UK
| |
Collapse
|
42
|
Zampino R, Iossa D, Ursi MP, Bertolino L, Karruli A, Molaro R, Esposito G, Vitrone M, D’Amico F, Albisinni R, Durante-Mangoni E. Clinical Significance and Prognostic Value of Hemostasis Parameters in 337 Patients with Acute Infective Endocarditis. J Clin Med 2021; 10:jcm10225386. [PMID: 34830670 PMCID: PMC8624946 DOI: 10.3390/jcm10225386] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 12/16/2022] Open
Abstract
(1) Background: The aim of this study was to assess the clinical significance and prognostic role of the main hemostasis parameters in infective endocarditis (IE): prothrombin time as international normalized ratio (PT-INR), activated partial thromboplastin time (aPTT), fibrinogen, D-dimers, platelet count, homocysteine. (2) Methods: We studied 337 patients with IE. Clinical, hemato-chemical and echocardiography parameters were analyzed. Coagulation parameters were measured on admission. (3) Results: D-dimers levels (p = 0.012) and a prolonged PT-INR (p = 0.013) were associated with higher in-hospital mortality, while prolonged aPTT (p = 0.021) was associated with increased 1-year mortality. Staphylococcus aureus (S. aureus) infection (p = 0.003), prosthetic valve endocarditis (PVE) (p = 0.001), surgical indication (p = 0.002) and higher D-dimer levels (p = 0.005) were independent predictors of in-hospital mortality. PVE (p = 0.001), a higher Charlson Comorbidity Index (p = 0.049), surgical indication (p = 0.001) and prolonged aPTT (p = 0.012) were independent predictors of 1-year mortality. Higher levels of D-dimers (p < 0.001) and a shorter aPTT (p < 0.001) were associated with embolic complications of IE. S. aureus etiology was bound to higher D-dimers levels (p < 0.001) and a shorter aPTT (p = 0.006). (4) Conclusions: Elevated D-dimers are associated with a higher risk for in-hospital mortality in IE patients. High D-dimers and a short aPTT are associated with a higher risk for embolic events in IE. A longer aPTT is associated with 1-year mortality.
Collapse
Affiliation(s)
- Rosa Zampino
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.Z.); (M.P.U.); (L.B.); (G.E.); (M.V.)
- Unit of Infectious & Transplant Medicine, A.O.R.N. Ospedali dei Colli-Ospedale Monaldi, 80131 Naples, Italy; (D.I.); (F.D.); (R.A.)
| | - Domenico Iossa
- Unit of Infectious & Transplant Medicine, A.O.R.N. Ospedali dei Colli-Ospedale Monaldi, 80131 Naples, Italy; (D.I.); (F.D.); (R.A.)
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (A.K.); (R.M.)
| | - Maria Paola Ursi
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.Z.); (M.P.U.); (L.B.); (G.E.); (M.V.)
| | - Lorenzo Bertolino
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.Z.); (M.P.U.); (L.B.); (G.E.); (M.V.)
| | - Arta Karruli
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (A.K.); (R.M.)
| | - Rosa Molaro
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (A.K.); (R.M.)
| | - Gennaro Esposito
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.Z.); (M.P.U.); (L.B.); (G.E.); (M.V.)
| | - Martina Vitrone
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (R.Z.); (M.P.U.); (L.B.); (G.E.); (M.V.)
| | - Fabiana D’Amico
- Unit of Infectious & Transplant Medicine, A.O.R.N. Ospedali dei Colli-Ospedale Monaldi, 80131 Naples, Italy; (D.I.); (F.D.); (R.A.)
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (A.K.); (R.M.)
| | - Rosina Albisinni
- Unit of Infectious & Transplant Medicine, A.O.R.N. Ospedali dei Colli-Ospedale Monaldi, 80131 Naples, Italy; (D.I.); (F.D.); (R.A.)
| | - Emanuele Durante-Mangoni
- Unit of Infectious & Transplant Medicine, A.O.R.N. Ospedali dei Colli-Ospedale Monaldi, 80131 Naples, Italy; (D.I.); (F.D.); (R.A.)
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (A.K.); (R.M.)
- Correspondence: ; Tel.: +39-(0)817062475; Fax: +39-(0)817702645
| | | |
Collapse
|
43
|
Leeten K, Jacques N, Lancellotti P, Oury C. Aspirin or Ticagrelor in Staphylococcus aureus Infective Endocarditis: Where Do We Stand? Front Cell Dev Biol 2021; 9:716302. [PMID: 34692677 PMCID: PMC8529053 DOI: 10.3389/fcell.2021.716302] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/21/2021] [Indexed: 12/04/2022] Open
Abstract
Infective endocarditis is a challenging disease with a high mortality and morbidity rate. Antibiotic prophylaxis is currently recommended in high-risk infective endocarditis patients. However, the use of antibiotics faces the challenge of a low efficacy and contributes further to the emerging infection rate by antibiotic-resistant strains, emphasizing the need for new therapeutic strategies. Platelets are essential in the initial phase of infective endocarditis, acting as first-line immune responders. During the first phase of disease, bacteria can interact with platelets and counteract platelet antimicrobial activities. Mechanistic in vitro and animal studies on the effect of aspirin on bacteria-platelet interactions and the prevention of vegetation development showed promising results. However, data from clinical studies on the outcome of infective endocarditis patients who were receiving medically indicated aspirin therapy remain controversial. Therefore, the benefit of antiplatelet agents in infective endocarditis prevention has been questioned. Besides aspirin, it has been discovered that the platelet P2Y12 receptor antagonist ticagrelor has antibacterial properties in addition to its potent antiplatelet activity. Furthermore, a recent study in mice and a case report remarkably indicated the ability of this drug to eradicate Staphylococcus aureus bacteremia. This review will focus on current knowledge on antibacterial activity of ticagrelor, compared to aspirin, pointing out main unanswered questions. The goal is to provide food for thought as to whether a prior ticagrelor therapy might be beneficial for the prevention of infective endocarditis.
Collapse
Affiliation(s)
- Kirsten Leeten
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, Liège, Belgium
| | - Nicolas Jacques
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, Liège, Belgium
| | - Patrizio Lancellotti
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, Liège, Belgium.,Gruppo Villa Maria Care and Research, Maria Cecilia Hospital, Cotignola, and Anthea Hospital, Bari, Italy
| | - Cécile Oury
- Laboratory of Cardiology, Department of Cardiology, GIGA Institute, University of Liège Hospital, Liège, Belgium
| |
Collapse
|
44
|
A Specific Host/Microbial Signature of Plasma-Derived Extracellular Vesicles Is Associated to Thrombosis and Marrow Fibrosis in Polycythemia Vera. Cancers (Basel) 2021; 13:cancers13194968. [PMID: 34638452 PMCID: PMC8507916 DOI: 10.3390/cancers13194968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Patients with polycythemia vera, a myeloproliferative neoplasm, are at increased risk of thrombosis and progression to myelofibrosis. However, no disease-specific risk factors have been identified so far. Extracellular vesicles, released from a broad variety of cells, are receiving increasing attention for their effects on cell-to-cell communication. In addition, they play a role in cancer and thrombosis. Interestingly, circulating microbial components/microbes have been recently indicated as potential modifiers of inflammation and coagulation. Here, we identified a signature of thrombosis history and marrow fibrosis by analyzing the phenotype and the microbial DNA cargo of the circulating extracellular vesicles after isolation from the plasma of patients with polycythemia vera. These data may support the role of extracellular vesicles as liquid biomarkers of aggressive disease, thus contributing to refining the prognosis of polycythemia vera. Abstract Polycythemia vera is a myeloproliferative neoplasm with increased risk of thrombosis and progression to myelofibrosis. However, no disease-specific risk factors have been identified so far. Circulating extracellular vesicles (EVs) are mostly of megakaryocyte (MK-EVs) and platelet (PLT-EVs) origin and, along with phosphatidylethanolamine (PE)-EVs, play a role in cancer and thrombosis. Interestingly, circulating microbial components/microbes have been recently indicated as potential modifiers of inflammation and coagulation. Here, we investigated phenotype and microbial DNA cargo of EVs after isolation from the plasma of 38 patients with polycythemia vera. Increased proportion of MK-EVs and reduced proportion of PLT-EVs identify patients with thrombosis history. Interestingly, EVs from patients with thrombosis history were depleted in Staphylococcus DNA but enriched in DNA from Actinobacteria members as well as Anaerococcus. In addition, patients with thrombosis history had also lower levels of lipopolysaccharide-associated EVs. In regard to fibrosis, along with increased proportion of PE-EVs, the EVs of patients with marrow fibrosis were enriched in DNA from Collinsella and Flavobacterium. Here, we identified a polycythemia-vera-specific host/microbial EV-based signature associated to thrombosis history and marrow fibrosis. These data may contribute to refining PV prognosis and to identifying novel druggable targets.
Collapse
|
45
|
Pezzanite LM, Hendrickson DA, Dow S, Stoneback J, Chow L, Krause D, Goodrich L. Intra-articular administration of antibiotics in horses: Justifications, risks, reconsideration of use and outcomes. Equine Vet J 2021; 54:24-38. [PMID: 34459027 DOI: 10.1111/evj.13502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 07/02/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022]
Abstract
Antibiotics have been injected intra-articularly by equine veterinarians for decades, either prophylactically when other drugs are administered for osteoarthritis or therapeutically to treat septic arthritis. This route of administration has also more recently gained attention in human orthopaedic clinical practice, particularly as an alternative to systemic antibiotic administration to treat infections following prosthetic arthroplasty. While the rationale for injecting antibiotics intra-articularly has been largely focused on achieving high local drug concentrations, there has been relatively little focus on pharmacokinetic parameters of antibiotics administered by this route, or on the potential for local toxicity. The increasing incidence of antibiotic resistance in veterinary and human medicine prompts reconsideration of off-label antibiotic usage and evaluation of evidence-based dosing strategies. The purpose of this review was to summarise the current literature describing intra-articular antibiotic usage, including specific studies where pharmacokinetics, potential safety and toxicity have been evaluated. This review will advance practitioners' understanding of the use of intra-articularly administered antibiotics, including the overall pros and cons of the approach.
Collapse
Affiliation(s)
- Lynn M Pezzanite
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Dean A Hendrickson
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Steven Dow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA.,Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Jason Stoneback
- Department of Orthopedic Surgery, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Lyndah Chow
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Danielle Krause
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Laurie Goodrich
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
46
|
Kearney KJ, Ariëns RAS, Macrae FL. The Role of Fibrin(ogen) in Wound Healing and Infection Control. Semin Thromb Hemost 2021; 48:174-187. [PMID: 34428799 DOI: 10.1055/s-0041-1732467] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Fibrinogen, one of the most abundant plasma proteins playing a key role in hemostasis, is an important modulator of wound healing and host defense against microbes. In the current review, we address the role of fibrin(ogen) throughout the process of wound healing and subsequent tissue repair. Initially fibrin(ogen) acts as a provisional matrix supporting incoming leukocytes and acting as reservoir for growth factors. It later goes on to support re-epithelialization, angiogenesis, and fibroplasia. Importantly, removal of fibrin(ogen) from the wound is essential for wound healing to progress. We also discuss how fibrin(ogen) functions through several mechanisms to protect the host against bacterial infection by providing a physical barrier, entrapment of bacteria in fibrin(ogen) networks, and by directing immune cell function. The central role of fibrin(ogen) in defense against bacterial infection has made it a target of bacterial proteins, evolved to interact with fibrin(ogen) to manipulate clot formation and degradation for the purpose of promoting microbial virulence and survival. Further understanding of the dual roles of fibrin(ogen) in wound healing and infection could provide novel means of therapy to improve recovery from surgical or chronic wounds and help to prevent infection from highly virulent bacterial strains, including those resistant to antibiotics.
Collapse
Affiliation(s)
- Katherine J Kearney
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Fraser L Macrae
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
47
|
Abstract
Staphylococcus aureus can target a variety of tissues, causing life-threatening infections. The basis for this diversity stems from the microorganism’s ability to spread in the vascular system throughout the body. To survive in blood, S. aureus coats itself with a fibrinogen (Fg)/fibrin shield. The protective shield is assembled by the coordinated actions of a number of Fg-binding bacterial proteins that manipulate the host’s blood coagulation system. Several of the Fg binders appear redundant, sharing similar functional motifs. This observation led us to screen for the presence of novel proteins with significant amino acid identities to von Willebrand factor-binding protein (vWbp), a key component in the shield assembly machinery. One identified protein showed significant sequence identity with the C-terminal region of vWbp, and we consequently named it vWbp homologous protein (vhp). The vhp gene lies within a cluster of genes that encode other virulence factors in S. aureus. Although each isolate only contains one copy of the vhp gene, S. aureus has at least three distinct alleles, vhpA, B, and C, that are present in the core genome. All three vhp isoforms bind Fg with high affinity, targeting a site located in the D fragment of Fg. We further identified an ∼79 amino acid-long, conserved segment within the C-terminal region of vWbp that shares high sequence identities (54 to 67%) with the vhps and binds soluble Fg with high affinity. Further analysis of this conserved motif and the intact vhps revealed intriguing differences in the Fg binding behavior, perhaps suggesting that these proteins have similar but discrete functions in the shield assembly.
Collapse
|
48
|
Silasi R, Keshari RS, Regmi G, Lupu C, Georgescu C, Simmons JH, Wallisch M, Kohs TCL, Shatzel JJ, Olson SR, Lorentz CU, Puy C, Tucker EI, Gailani D, Strickland S, Gruber A, McCarty OJT, Lupu F. Factor XII plays a pathogenic role in organ failure and death in baboons challenged with Staphylococcus aureus. Blood 2021; 138:178-189. [PMID: 33598692 PMCID: PMC8288658 DOI: 10.1182/blood.2020009345] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/26/2021] [Indexed: 12/29/2022] Open
Abstract
Activation of coagulation factor (F) XI promotes multiorgan failure in rodent models of sepsis and in a baboon model of lethal systemic inflammation induced by infusion of heat-inactivated Staphylococcus aureus. Here we used the anticoagulant FXII-neutralizing antibody 5C12 to verify the mechanistic role of FXII in this baboon model. Compared with untreated control animals, repeated 5C12 administration before and at 8 and 24 hours after bacterial challenge prevented the dramatic increase in circulating complexes of contact system enzymes FXIIa, FXIa, and kallikrein with antithrombin or C1 inhibitor, and prevented cleavage and consumption of high-molecular-weight kininogen. Activation of several coagulation factors and fibrinolytic enzymes was also prevented. D-dimer levels exhibited a profound increase in the untreated animals but not in the treated animals. The antibody also blocked the increase in plasma biomarkers of inflammation and cell damage, including tumor necrosis factor, interleukin (IL)-1β, IL-6, IL-8, IL-10, granulocyte-macrophage colony-stimulating factor, nucleosomes, and myeloperoxidase. Based on clinical presentation and circulating biomarkers, inhibition of FXII prevented fever, terminal hypotension, respiratory distress, and multiorgan failure. All animals receiving 5C12 had milder and transient clinical symptoms and were asymptomatic at day 7, whereas untreated control animals suffered irreversible multiorgan failure and had to be euthanized within 2 days after the bacterial challenge. This study confirms and extends our previous finding that at least 2 enzymes of the contact activation complex, FXIa and FXIIa, play critical roles in the development of an acute and terminal inflammatory response in baboons challenged with heat-inactivated S aureus.
Collapse
Affiliation(s)
- Robert Silasi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Ravi S Keshari
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Girija Regmi
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Cristina Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Constantin Georgescu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Joe H Simmons
- Michale E. Keeling Center for Comparative Medicine and Research, University of Texas MD Anderson Cancer Center, Bastrop, TX
| | - Michael Wallisch
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR
- Aronora, Inc, Portland, OR
| | - Tia C L Kohs
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR
| | - Joseph J Shatzel
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR
- Division of Hematology & Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR
| | - Sven R Olson
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR
- Division of Hematology & Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR
| | - Christina U Lorentz
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR
- Aronora, Inc, Portland, OR
| | - Cristina Puy
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR
| | - Erik I Tucker
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR
- Aronora, Inc, Portland, OR
| | - David Gailani
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY; and
| | - András Gruber
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR
- Aronora, Inc, Portland, OR
- Division of Hematology & Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR
| | - Owen J T McCarty
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR
- Division of Hematology & Medical Oncology, School of Medicine, Oregon Health & Science University, Portland, OR
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Cell Biology
- Department of Pathology, and
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
49
|
Karczewski J, Brown CM, Maezato Y, Krasucki SP, Streatfield SJ. Efficacy of a novel lantibiotic, CMB001, against MRSA. J Antimicrob Chemother 2021; 76:1532-1538. [PMID: 33582800 DOI: 10.1093/jac/dkab040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/22/2021] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES To evaluate the efficacy of a novel lantibiotic, CMB001, against MRSA biofilms in vitro and in an in vivo experimental model of bacterial infection. METHODS Antibacterial activity of CMB001 was measured in vitro after its exposure to whole blood or to platelet-poor plasma. In vitro efficacy of CMB001 against a Staphylococcus aureus biofilm was studied using scanning electron microscopy. The maximum tolerable dose in mice was determined and a preliminary pharmacokinetic analysis for CMB001 was performed in mice. In vivo efficacy was evaluated in a neutropenic mouse thigh model of infection. RESULTS CMB001 maintained its antibacterial activity in the presence of blood or plasma for up to 24 h at 37°C. CMB001 efficiently killed S. aureus within the biofilm by causing significant damage to the bacterial cell wall. The maximum tolerable dose in mice was established to be 10 mg/kg and could be increased to 30 mg/kg in mice pretreated with antihistamines. In neutropenic mice infected with MRSA, treatment with CMB001 reduced the bacterial burden with an efficacy equivalent to that of vancomycin. CONCLUSIONS CMB001 offers potential as an alternative treatment option to combat MRSA. It will be of interest to evaluate the in vivo efficacy of CMB001 against infections caused by other pathogens, including Clostridioides difficile and Acinetobacter baumannii, and to expand its pharmacokinetic/pharmacodynamic parameters and safety profile.
Collapse
Affiliation(s)
- Jerzy Karczewski
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE 19711, USA
| | - Christine M Brown
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE 19711, USA
| | - Yukari Maezato
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE 19711, USA
| | - Stephen P Krasucki
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE 19711, USA
| | | |
Collapse
|
50
|
Larsen JB, Hvas AM. Fibrinolytic Alterations in Sepsis: Biomarkers and Future Treatment Targets. Semin Thromb Hemost 2021; 47:589-600. [PMID: 33878784 DOI: 10.1055/s-0041-1725096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sepsis is a life-threatening condition which develops as a dysregulated immune response in the face of infection and which is associated with profound hemostatic disturbances and in the most extreme cases disseminated intravascular coagulation (DIC). In addition, the fibrinolytic system is subject to alterations during infection and sepsis, and impaired fibrinolysis is currently considered a key player in sepsis-related microthrombus formation and DIC. However, we still lack reliable biomarkers to assess fibrinolysis in the clinical setting. Furthermore, drugs targeting the fibrinolytic system have potential value in sepsis patients with severe fibrinolytic disturbances, but these are still being tested in the preclinical stage. The present review provides an overview of key fibrinolytic changes in sepsis, reviews the current literature on potential laboratory markers of altered fibrinolysis in adult sepsis patients, and discusses future perspectives for diagnosis and treatment of fibrinolytic disturbances in sepsis patients.
Collapse
Affiliation(s)
- Julie Brogaard Larsen
- Thrombosis and Haemostasis Research Unit, Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Anne-Mette Hvas
- Thrombosis and Haemostasis Research Unit, Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|