1
|
Elmeazawy R, Sorour EAAF, Badreldeen AS. Effects of community-acquired pneumonia on biventricular cardiac functions in children. Ital J Pediatr 2025; 51:126. [PMID: 40247416 PMCID: PMC12007363 DOI: 10.1186/s13052-025-01965-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/27/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND This study aimed to evaluate the effects of community-acquired pneumonia (CAP) on cardiac function in children and compare the effectiveness of tissue Doppler imaging (TDI) and two-dimensional speckle tracking echocardiography (2D-STE) with conventional echocardiography in the early detection of biventricular cardiac dysfunction in children with CAP. METHODS The study included 50 hospitalized children diagnosed with CAP and 50 matched healthy controls. All patients underwent cardiac evaluation including conventional echocardiography, TDI, and 2D-STE. RESULTS Fifty children with CAP with a mean age of 5.02 ± 2.46 years participated in the study. Thirty-two were male (64.0%). LV systolic function (S) and RV diastolic function (E'/A') were significantly lower in the diseased group compared to the control group (p < 0.001). The myocardial performance index (MPI) of both ventricles was significantly higher in the diseased group compared to the control group (p < 0.001). In patients with CAP, the mean value of two-dimensional longitudinal strain (2D LS) was significantly lower than that of the control group (p < 0.001). No statistically significant differences in echocardiographic parameters were observed when comparing complicated and non-complicated CAP subgroups. CONCLUSION TDI and 2D-STE demonstrated the ability to detect early biventricular dysfunction in pediatric patients diagnosed with CAP.
Collapse
Affiliation(s)
- Rehab Elmeazawy
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta, Egypt.
| | | | | |
Collapse
|
2
|
Chong SY, Lew SQ, Alam T, Gaulke CA, Lau GW. Comparative analysis of the Streptococcus pneumoniae competence development in vitro versus in vivo during pneumonia-derived sepsis. Front Microbiol 2025; 16:1540511. [PMID: 39935640 PMCID: PMC11811101 DOI: 10.3389/fmicb.2025.1540511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025] Open
Abstract
Introduction The Streptococcus pneumoniae (pneumococcus) competence regulon is well-known for regulating genetic transformation but is also important for virulence. Some pneumococcal strains can enter a transient competent state for genetic transformation in an optimized competence-inducing medium when the threshold level of the peptide pheromone competence stimulating peptide is attained; upregulating the expression of three distinct phases of "early", "late" and "delayed" competence genes. Recently, we discovered that pneumococcus can naturally enter a prolonged competent state during acute pneumonia in mice. However, mechanisms driving competence development during host infection are rarely examined, and a direct comparison between in vitro and in vivo competence induction has not been performed. Methods We conducted a comparative gene expression analysis of pneumococcal competence development in vitro versus in vivo during pneumonia-derived sepsis in mice. We examined existing RNA-Seq data and performed validation using RNA obtained from an independent replicate experiment. Results and discussion Our analysis revealed both similarities and differences in the expression of "early", "late", and "delayed" competence between in vitro versus during pneumonia-derived sepsis. Our results may reveal new aspects of pneumococcal competence biology.
Collapse
Affiliation(s)
- Sook Yin Chong
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Shi Qian Lew
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Tauqeer Alam
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Christopher A Gaulke
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Gee W Lau
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
3
|
Campos-Rodríguez F, Chiner E, de la Rosa-Carrillo D, García-Cosío B, Hernádez-Hernández JR, Jiménez D, Méndez R, Molina-Molina M, Soto-Campos JG, Vaquero JM, Gonzalez-Barcala FJ. Respiratory Pathology and Cardiovascular Diseases: A Scoping Review. OPEN RESPIRATORY ARCHIVES 2025; 7:100392. [PMID: 39758960 PMCID: PMC11696865 DOI: 10.1016/j.opresp.2024.100392] [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: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 01/07/2025] Open
Abstract
Respiratory diseases and cardiovascular diseases (CVDs) have high prevalence and share common risk factors. In some respiratory diseases such as sleep apnoea and COPD, the evidence of their negative impact on the prognosis of CVDs seems clear. However, in other diseases it is less evident whether there is any direct relationship. With this in mind, our objective was to provide information that may be helpful to better understand the relationship between respiratory pathology and CVDs. There are different reasons for this relationship, such as shared risk factors, common pathophysiological mechanisms, side effects of treatment and the direct effect in the heart and great vessels of respiratory diseases. Indeed, aging and smoking are risk factors for CVDs and also for respiratory diseases such as obstructive sleep apnea (OSA), COPD and interstitial lung diseases (ILD). Furthermore, there are common pathophysiological mechanisms that affect both respiratory diseases and CVDs, such as accelerated atherosclerosis, microvascular dysfunction, endothelial dysfunction, inflammation, hypoxemia and oxidative stress. Besides that, it is well known that lung cancer, sarcoidosis and amyloidosis may directly affect the heart and great vessels. Finally, side effects of drugs for respiratory diseases and the discontinuation of treatments that are necessary for CVDs, such as β-blockers and aspirin, may have a deleterious impact on the cardiovascular system. In conclusion, the coexistence of respiratory diseases and CVDs is very common. It makes modifying diagnostic and therapeutic management necessary and is also a relevant prognostic factor.
Collapse
Affiliation(s)
- Francisco Campos-Rodríguez
- Respiratory Department, Hospital Universitario de Valme, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Eusebi Chiner
- Respiratory Department, Hospital Universitario of San Juan of Alicante, Alicante, Spain
| | | | - Borja García-Cosío
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Respiratory Department, Hospital Son Espases-IdISBa, Palma de Mallorca, Spain
| | | | - David Jiménez
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Respiratory Department, Ramón y Cajal Hospital and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- Medicine Department, University of Alcalá, Madrid, Spain
| | - Raúl Méndez
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Respiratory Department, La Fe University and Polytechnic Hospital, Valencia, Spain
- Respiratory Infections, Health Research Institute La Fe (IISLAFE), Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
| | - María Molina-Molina
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Interstitial Lung Disease (ILD) Unit, Respiratory Department, University Hospital of Bellvitge, IDIBELL, UB, Barcelona, Spain
| | | | - José-Manuel Vaquero
- Department of Pulmonary Medicine and Lung Transplantation, University Hospital Reina Sofia, Avenida Menendez Pidal s/n, 14004 Cordoba, Spain
| | - Francisco-Javier Gonzalez-Barcala
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Translational Research In Airway Diseases Group (TRIAD), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Respiratory Department, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
4
|
Parveen S, Bhat CV, Sagilkumar AC, Aziz S, Arya J, Dutta A, Dutta S, Show S, Sharma K, Rakshit S, Johnson JB, Nongthomba U, Banerjee A, Subramanian K. Bacterial pore-forming toxin pneumolysin drives pathogenicity through host extracellular vesicles released during infection. iScience 2024; 27:110589. [PMID: 39211544 PMCID: PMC11357855 DOI: 10.1016/j.isci.2024.110589] [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: 02/02/2024] [Revised: 06/11/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Streptococcus pneumoniae is a global priority respiratory pathogen that kills over a million people annually. The pore-forming cytotoxin, pneumolysin (PLY) is a major virulence factor. Here, we found that recombinant PLY as well as wild-type pneumococcal strains, but not the isogenic PLY mutant, upregulated the shedding of extracellular vesicles (EVs) harboring membrane-bound toxin from human THP-1 monocytes. PLY-EVs induced cytotoxicity and hemolysis dose-dependently upon internalization by recipient monocyte-derived dendritic cells. Proteomics analysis revealed that PLY-EVs are selectively enriched in key inflammatory host proteins such as IFI16, NLRC4, PTX3, and MMP9. EVs shed from PLY-challenged or infected cells induced dendritic cell maturation and primed them to infection. In vivo, zebrafish administered with PLY-EVs showed pericardial edema and mortality. Adoptive transfer of bronchoalveolar-lavage-derived EVs from infected mice to healthy recipients induced lung damage and inflammation in a PLY-dependent manner. Our findings identify that host EVs released during infection mediate pneumococcal pathogenesis.
Collapse
Affiliation(s)
- Saba Parveen
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Chinmayi V Bhat
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Aswathy C Sagilkumar
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
- Regional Centre for Biotechnology, Faridabad 121001, India
| | - Shaheena Aziz
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - J Arya
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Asmita Dutta
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Somit Dutta
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Sautan Show
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Kuldeep Sharma
- Bacterial Pathogenesis Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Sumit Rakshit
- Bacterial Pathogenesis Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - John Bernet Johnson
- Virology Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Upendra Nongthomba
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Anirban Banerjee
- Bacterial Pathogenesis Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Karthik Subramanian
- Host-Pathogen Laboratory, Pathogen Biology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
- Regional Centre for Biotechnology, Faridabad 121001, India
| |
Collapse
|
5
|
Sanford TC, Tweten RK, Abrahamsen HL. Bacterial cholesterol-dependent cytolysins and their interaction with the human immune response. Curr Opin Infect Dis 2024; 37:164-169. [PMID: 38527455 PMCID: PMC11042984 DOI: 10.1097/qco.0000000000001010] [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] [Indexed: 03/27/2024]
Abstract
PURPOSE OF REVIEW Many cholesterol-dependent cytolysin (CDC)-producing pathogens pose a significant threat to human health. Herein, we review the pore-dependent and -independent properties CDCs possess to assist pathogens in evading the host immune response. RECENT FINDINGS Within the last 5 years, exciting new research suggests CDCs can act to inhibit important immune functions, disrupt critical cell signaling pathways, and have tissue-specific effects. Additionally, recent studies have identified a key region of CDCs that generates robust immunity, providing resources for the development of CDC-based vaccines. SUMMARY This review provides new information on how CDCs alter host immune responses to aid bacteria in pathogenesis. These studies can assist in the design of more efficient vaccines and therapeutics against CDCs that will enhance the immune response to CDC-producing pathogens while mitigating the dampening effects CDCs have on the host immune response.
Collapse
Affiliation(s)
- Tristan C. Sanford
- University of Oklahoma Health Sciences Center, Department of Microbiology and Immunology, Oklahoma City, OK 73104
| | - Rodney K. Tweten
- University of Oklahoma Health Sciences Center, Department of Microbiology and Immunology, Oklahoma City, OK 73104
| | - Hunter L. Abrahamsen
- University of Oklahoma Health Sciences Center, Department of Microbiology and Immunology, Oklahoma City, OK 73104
| |
Collapse
|
6
|
Oh MW, Lin J, Chong SY, Lew SQ, Alam T, Lau GW. Time-resolved RNA-seq analysis to unravel the in vivo competence induction by Streptococcus pneumoniae during pneumonia-derived sepsis. Microbiol Spectr 2024; 12:e0305023. [PMID: 38305162 PMCID: PMC10913500 DOI: 10.1128/spectrum.03050-23] [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: 08/08/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
Competence development in Streptococcus pneumoniae (pneumococcus) is tightly intertwined with virulence. In addition to genes encoding genetic transformation machinery, the competence regulon also regulates the expression of allolytic factors, bacteriocins, and cytotoxins. Pneumococcal competence system has been extensively interrogated in vitro where the short transient competent state upregulates the expression of three distinct phases of "early," "late," and "delayed" genes. Recently, we have demonstrated that the pneumococcal competent state develops naturally in mouse models of pneumonia-derived sepsis. To unravel the underlying adaptive mechanisms driving the development of the competent state, we conducted a time-resolved transcriptomic analysis guided by the spatiotemporal live in vivo imaging system of competence induction during pneumonia-derived sepsis. Mouse lungs infected by the serotype 2 strain D39 expressing a competent state-specific reporter gene (D39-ssbB-luc) were subjected to RNA sequencing guided by monitoring the competence development at 0, 12, 24, and, at the moribund state, >40 hours post-infection (hpi). Transcriptomic analysis revealed that the competence-specific gene expression patterns in vivo were distinct from those under in vitro conditions. There was significant upregulation of early, late, and some delayed phase competence-specific genes as early as 12 hpi, suggesting that the pneumococcal competence regulon is important for adaptation to the lung environment. Additionally, members of the histidine triad (pht) gene family were sharply upregulated at 12 hpi followed by a steep decline throughout the rest of the infection cycle, suggesting that Pht proteins participate in the early adaptation to the lung environment. Further analysis revealed that Pht proteins execute a metal ion-dependent regulatory role in competence induction.IMPORTANCEThe induction of pneumococcal competence for genetic transformation has been extensively studied in vitro but poorly understood during lung infection. We utilized a combination of live imaging and RNA sequencing to monitor the development of a competent state during acute pneumonia. Upregulation of competence-specific genes was observed as early as 12 hour post-infection, suggesting that the pneumococcal competence regulon plays an important role in adapting pneumococcus to the stressful lung environment. Among others, we report novel finding that the pneumococcal histidine triad (pht) family of genes participates in the adaptation to the lung environment and regulates pneumococcal competence induction.
Collapse
Affiliation(s)
- Myung Whan Oh
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jingjun Lin
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Sook Yin Chong
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Shi Qian Lew
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Tauqeer Alam
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Gee W. Lau
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
7
|
Zou Y, Wang H, Fang J, Sun H, Deng X, Wang J, Deng Y, Chi G. Isorhamnetin as a novel inhibitor of pneumolysin against Streptococcus pneumoniae infection in vivo/in vitro. Microb Pathog 2023; 185:106382. [PMID: 37839759 DOI: 10.1016/j.micpath.2023.106382] [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: 04/10/2023] [Revised: 09/18/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023]
Abstract
The increasing incidence of Streptococcus pneumoniae (S. pneumoniae) infection severely threatened the global public heath, causing a significant fatality in immunocompromised hosts. Notably, pneumolysin (PLY) as a pore-forming cytolysin plays a crucial role in the pathogenesis of pneumococcal pneumonia and lung injury. In this study, a natural flavonoid isorhamnetin was identified as a PLY inhibition to suppress PLY-induced hemolysis by engaging the predicted residues and attenuate cytolysin PLY-mediated A549 cells injury. Underlying mechanisms revealed that PLY inhibitor isorhamnetin further contributed to decrease the formation of bacterial biofilms without affecting the expression of PLY. In vivo S. pneumoniae infection confirmed that the pathological injury of lung tissue evoked by S. pneumoniae was ameliorated by isorhamnetin treatment. Collectively, these results presented that isorhamnetin could inhibit the biological activity of PLY, thus reducing the pathogenicity of S. pneumoniae. In summary, our study laid a foundation for the feasible anti-virulence strategy targeting PLY, and provided a promising PLY inhibitor for the treatment of S. pneumoniae infection.
Collapse
Affiliation(s)
- Yinuo Zou
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Haiting Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Juan Fang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hongxiang Sun
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Xuming Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianfeng Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yanhong Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Gefu Chi
- The Affiliated Hospital of Inner Mongolia Medical University, Huhhot, Nei Monggol, China.
| |
Collapse
|
8
|
Taha AM, Mahmoud AM, Ghonaim MM, Kamran A, AlSamhori JF, AlBarakat MM, Shrestha AB, Jaiswal V, Reiter RJ. Melatonin as a potential treatment for septic cardiomyopathy. Biomed Pharmacother 2023; 166:115305. [PMID: 37619482 DOI: 10.1016/j.biopha.2023.115305] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
Septic cardiomyopathy (SCM) is a common complication of sepsis contributing to high mortality rates. Its pathophysiology involves complex factors, including inflammatory cytokines, mitochondrial dysfunction, oxidative stress, and immune dysregulation. Despite extensive research, no effective pharmacological agent has been established for sepsis-induced cardiomyopathy. Melatonin, a hormone with diverse functions in the body, has emerged as a potential agent for SCM through its anti-oxidant, anti-inflammatory, anti-apoptotic, and cardioprotective roles. Through various molecular levels of its mechanism of action, it counterattacks the adverse event of sepsis. Experimental studies have mentioned that melatonin protects against many cardiovascular diseases and exerts preventive effects on SCM. Moreover, melatonin has been investigated in combination with other drugs such as antibiotics, resveratrol, and anti-oxidants showing synergistic effects in reducing inflammation, anti-oxidant, and improving cardiac function. While preclinical studies have demonstrated positive results, clinical trials are required to establish the optimal dosage, route of administration, and treatment duration for melatonin in SCM. Its safety profile, low toxicity, and natural occurrence in the human body provide a favorable basis for its clinical use. This review aims to provide an overview of the current evidence of the use of melatonin in sepsis-induced cardiomyopathy (SICM). Melatonin appears to be promising as a possible treatment for sepsis-induced cardiomyopathy and demands further investigation.
Collapse
Affiliation(s)
- Amira Mohamed Taha
- Faculty of Medicine, Fayoum University, Fayoum, Egypt; Medical Research Group of Egypt (MRGE), Negida Academy, Arlington, MA, USA
| | | | | | - Ateeba Kamran
- Bachelor of Medicine, Bachelor of Surgery, Karachi Medical and Dental College, Karachi, Pakistan
| | | | - Majd M AlBarakat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Abhigan Babu Shrestha
- Department of Internal Medicine, M Abdur Rahim Medical College, Dinajpur, Bangladesh.
| | | | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
9
|
Ferreira G, Cardozo R, Sastre S, Costa C, Santander A, Chavarría L, Guizzo V, Puglisi J, Nicolson GL. Bacterial toxins and heart function: heat-labile Escherichia coli enterotoxin B promotes changes in cardiac function with possible relevance for sudden cardiac death. Biophys Rev 2023; 15:447-473. [PMID: 37681088 PMCID: PMC10480140 DOI: 10.1007/s12551-023-01100-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/11/2023] [Indexed: 09/09/2023] Open
Abstract
Bacterial toxins can cause cardiomyopathy, though it is not its most common cause. Some bacterial toxins can form pores in the membrane of cardiomyocytes, while others can bind to membrane receptors. Enterotoxigenic E. coli can secrete enterotoxins, including heat-resistant (ST) or labile (LT) enterotoxins. LT is an AB5-type toxin that can bind to specific cell receptors and disrupt essential host functions, causing several common conditions, such as certain diarrhea. The pentameric B subunit of LT, without A subunit (LTB), binds specifically to certain plasma membrane ganglioside receptors, found in lipid rafts of cardiomyocytes. Isolated guinea pig hearts and cardiomyocytes were exposed to different concentrations of purified LTB. In isolated hearts, mechanical and electrical alternans and an increment of heart rate variability, with an IC50 of ~0.2 μg/ml LTB, were observed. In isolated cardiomyocytes, LTB promoted significant decreases in the amplitude and the duration of action potentials. Na+ currents were inhibited whereas L-type Ca2+ currents were augmented at their peak and their fast inactivation was promoted. Delayed rectifier K+ currents decreased. Measurements of basal Ca2+ or Ca2+ release events in cells exposed to LTB suggest that LTB impairs Ca2+ homeostasis. Impaired calcium homeostasis is linked to sudden cardiac death. The results are consistent with the recent view that the B subunit is not merely a carrier of the A subunit, having a role explaining sudden cardiac death in children (SIDS) infected with enterotoxigenic E. coli, explaining several epidemiological findings that establish a strong relationship between SIDS and ETEC E. coli. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-023-01100-6.
Collapse
Affiliation(s)
- Gonzalo Ferreira
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Romina Cardozo
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Santiago Sastre
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics and Centro de Investigaciones Biomédicas (CeInBio), Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Carlos Costa
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Axel Santander
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Luisina Chavarría
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - Valentina Guizzo
- Ion Channels, Biological Membranes and Cell Signaling Laboratory, Dept. Of Biophysics, Facultad de Medicina, Universidad de la Republica, Gral Flores 2125, 11800 Montevideo, CP Uruguay
| | - José Puglisi
- College of Medicine, California North State University, 9700 West Taron Drive, Elk Grove, CA 95757 USA
| | - G. L. Nicolson
- Institute for Molecular Medicine, Beach, Huntington, CA USA
| |
Collapse
|
10
|
Stotts C, Corrales-Medina VF, Rayner KJ. Pneumonia-Induced Inflammation, Resolution and Cardiovascular Disease: Causes, Consequences and Clinical Opportunities. Circ Res 2023; 132:751-774. [PMID: 36927184 DOI: 10.1161/circresaha.122.321636] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Pneumonia is inflammation in the lungs, which is usually caused by an infection. The symptoms of pneumonia can vary from mild to life-threatening, where severe illness is often observed in vulnerable populations like children, older adults, and those with preexisting health conditions. Vaccines have greatly reduced the burden of some of the most common causes of pneumonia, and the use of antimicrobials has greatly improved the survival to this infection. However, pneumonia survivors do not return to their preinfection health trajectories but instead experience an accelerated health decline with an increased risk of cardiovascular disease. The mechanisms of this association are not well understood, but a persistent dysregulated inflammatory response post-pneumonia appears to play a central role. It is proposed that the inflammatory response during pneumonia is left unregulated and exacerbates atherosclerotic vascular disease, which ultimately leads to adverse cardiac events such as myocardial infarction. For this reason, there is a need to better understand the inflammatory cross talk between the lungs and the heart during and after pneumonia to develop therapeutics that focus on preventing pneumonia-associated cardiovascular events. This review will provide an overview of the known mechanisms of inflammation triggered during pneumonia and their relevance to the increased cardiovascular risk that follows this infection. We will also discuss opportunities for new clinical approaches leveraging strategies to promote inflammatory resolution pathways as a novel therapeutic target to reduce the risk of cardiac events post-pneumonia.
Collapse
Affiliation(s)
- Cameron Stotts
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., K.J.R).,Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., V.F.C.-M.).,University of Ottawa Heart Institute, Ottawa, ON, Canada (C.S., K.J.R)
| | - Vicente F Corrales-Medina
- Centre for Infection, Immunity, and Inflammation, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., V.F.C.-M.).,Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (V.F.C-M).,Ottawa Hospital Research Institute, Ottawa, ON, Canada (V.F.C.-M)
| | - Katey J Rayner
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada (C.S., K.J.R).,University of Ottawa Heart Institute, Ottawa, ON, Canada (C.S., K.J.R)
| |
Collapse
|
11
|
Kruckow KL, Zhao K, Bowdish DME, Orihuela CJ. Acute organ injury and long-term sequelae of severe pneumococcal infections. Pneumonia (Nathan) 2023; 15:5. [PMID: 36870980 PMCID: PMC9985869 DOI: 10.1186/s41479-023-00110-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/31/2023] [Indexed: 03/06/2023] Open
Abstract
Streptococcus pneumoniae (Spn) is a major public health problem, as it is a main cause of otitis media, community-acquired pneumonia, bacteremia, sepsis, and meningitis. Acute episodes of pneumococcal disease have been demonstrated to cause organ damage with lingering negative consequences. Cytotoxic products released by the bacterium, biomechanical and physiological stress resulting from infection, and the corresponding inflammatory response together contribute to organ damage accrued during infection. The collective result of this damage can be acutely life-threatening, but among survivors, it also contributes to the long-lasting sequelae of pneumococcal disease. These include the development of new morbidities or exacerbation of pre-existing conditions such as COPD, heart disease, and neurological impairments. Currently, pneumonia is ranked as the 9th leading cause of death, but this estimate only considers short-term mortality and likely underestimates the true long-term impact of disease. Herein, we review the data that indicates damage incurred during acute pneumococcal infection can result in long-term sequelae which reduces quality of life and life expectancy among pneumococcal disease survivors.
Collapse
Affiliation(s)
- Katherine L Kruckow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kevin Zhao
- McMaster Immunology Research Centre and the Firestone Institute for Respiratory Health, McMaster University, Hamilton, Canada
| | - Dawn M E Bowdish
- McMaster Immunology Research Centre and the Firestone Institute for Respiratory Health, McMaster University, Hamilton, Canada
| | - Carlos J Orihuela
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
12
|
Abrams ST, Alhamdi Y, Zi M, Guo F, Du M, Wang G, Cartwright EJ, Toh CH. Extracellular Histone-Induced Protein Kinase C Alpha Activation and Troponin Phosphorylation Is a Potential Mechanism of Cardiac Contractility Depression in Sepsis. Int J Mol Sci 2023; 24:ijms24043225. [PMID: 36834636 PMCID: PMC9967552 DOI: 10.3390/ijms24043225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/29/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Reduction in cardiac contractility is common in severe sepsis. However, the pathological mechanism is still not fully understood. Recently it has been found that circulating histones released after extensive immune cell death play important roles in multiple organ injury and disfunction, particularly in cardiomyocyte injury and contractility reduction. How extracellular histones cause cardiac contractility depression is still not fully clear. In this work, using cultured cardiomyocytes and a histone infusion mouse model, we demonstrate that clinically relevant histone concentrations cause significant increases in intracellular calcium concentrations with subsequent activation and enriched localization of calcium-dependent protein kinase C (PKC) α and βII into the myofilament fraction of cardiomyocytes in vitro and in vivo. Furthermore, histones induced dose-dependent phosphorylation of cardiac troponin I (cTnI) at the PKC-regulated phosphorylation residues (S43 and T144) in cultured cardiomyocytes, which was also confirmed in murine cardiomyocytes following intravenous histone injection. Specific inhibitors against PKCα and PKCβII revealed that histone-induced cTnI phosphorylation was mainly mediated by PKCα activation, but not PKCβII. Blocking PKCα also significantly abrogated histone-induced deterioration in peak shortening, duration and the velocity of shortening, and re-lengthening of cardiomyocyte contractility. These in vitro and in vivo findings collectively indicate a potential mechanism of histone-induced cardiomyocyte dysfunction driven by PKCα activation with subsequent enhanced phosphorylation of cTnI. These findings also indicate a potential mechanism of clinical cardiac dysfunction in sepsis and other critical illnesses with high levels of circulating histones, which holds the potential translational benefit to these patients by targeting circulating histones and downstream pathways.
Collapse
Affiliation(s)
- Simon T. Abrams
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Coagulation Department, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK
| | - Yasir Alhamdi
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Sheffield Teaching Hospital NHS Foundation Trust, Sheffield S5 7AU, UK
| | - Min Zi
- Institute of Cardiovascular Sciences, Centre for Cardiac Research, University of Manchester, Manchester M13 9PT, UK
| | - Fengmei Guo
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- The Medical School, Southeast University, Nanjing 210009, China
| | - Min Du
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
| | - Guozheng Wang
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Coagulation Department, Liverpool University Hospitals NHS Foundation Trust, Liverpool L7 8XP, UK
- Correspondence: (G.W.); (C.-H.T.)
| | - Elizabeth J. Cartwright
- Institute of Cardiovascular Sciences, Centre for Cardiac Research, University of Manchester, Manchester M13 9PT, UK
| | - Cheng-Hock Toh
- Department of Clinical Infection Microbiology and Immunology, University of Liverpool, Liverpool L69 7BE, UK
- Roald Dahl Haemostasis & Thrombosis Centre, Royal Liverpool University Hospital, Liverpool L7 8XP, UK
- Correspondence: (G.W.); (C.-H.T.)
| |
Collapse
|
13
|
Cima Cabal MD, Molina F, López-Sánchez JI, Pérez-Santín E, Del Mar García-Suárez M. Pneumolysin as a target for new therapies against pneumococcal infections: A systematic review. PLoS One 2023; 18:e0282970. [PMID: 36947540 PMCID: PMC10032530 DOI: 10.1371/journal.pone.0282970] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/28/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND This systematic review evaluates pneumolysin (PLY) as a target for new treatments against pneumococcal infections. Pneumolysin is one of the main virulence factors produced by all types of pneumococci. This toxin (53 kDa) is a highly conserved protein that binds to cholesterol in eukaryotic cells, forming pores that lead to cell destruction. METHODS The databases consulted were MEDLINE, Web of Science, and Scopus. Articles were independently screened by title, abstract, and full text by two researchers, and using consensus to resolve any disagreements that occurred. Articles in other languages different from English, patents, cases report, notes, chapter books and reviews were excluded. Searches were restricted to the years 2000 to 2021. Methodological quality was evaluated using OHAT framework. RESULTS Forty-one articles describing the effects of different molecules that inhibit PLY were reviewed. Briefly, the inhibitory molecules found were classified into three main groups: those exerting a direct effect by binding and/or blocking PLY, those acting indirectly by preventing its effects on host cells, and those whose mechanisms are unknown. Although many molecules are proposed as toxin blockers, only some of them, such as antibiotics, peptides, sterols, and statins, have the probability of being implemented as clinical treatment. In contrast, for other molecules, there are limited studies that demonstrate efficacy in animal models with sufficient reliability. DISCUSSION Most of the studies reviewed has a good level of confidence. However, one of the limitations of this systematic review is the lack of homogeneity of the studies, what prevented to carry out a statistical comparison of the results or meta-analysis. CONCLUSION A panel of molecules blocking PLY activity are associated with the improvement of the inflammatory process triggered by the pneumococcal infection. Some molecules have already been used in humans for other purposes, so they could be safe for use in patients with pneumococcal infections. These patients might benefit from a second line treatment during the initial stages of the infection preventing acute respiratory distress syndrome and invasive pneumococcal diseases. Additional research using the presented set of compounds might further improve the clinical management of these patients.
Collapse
Affiliation(s)
- María Dolores Cima Cabal
- Escuela Superior de Ingeniería y Tecnología (ESIT), Universidad Internacional de La Rioja, UNIR, Logroño, La Rioja, Spain
| | - Felipe Molina
- Genética, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - José Ignacio López-Sánchez
- Escuela Superior de Ingeniería y Tecnología (ESIT), Universidad Internacional de La Rioja, UNIR, Logroño, La Rioja, Spain
| | - Efrén Pérez-Santín
- Escuela Superior de Ingeniería y Tecnología (ESIT), Universidad Internacional de La Rioja, UNIR, Logroño, La Rioja, Spain
| | - María Del Mar García-Suárez
- Escuela Superior de Ingeniería y Tecnología (ESIT), Universidad Internacional de La Rioja, UNIR, Logroño, La Rioja, Spain
| |
Collapse
|
14
|
The Importance of Pore-Forming Toxins in Multiple Organ Injury and Dysfunction. Biomedicines 2022; 10:biomedicines10123256. [PMID: 36552012 PMCID: PMC9776026 DOI: 10.3390/biomedicines10123256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/03/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Multiple organ injury and dysfunction often occurs in acute critical illness and adversely affects survival. However, in patients who survive, organ function usually recovers without permanent damage. It is, therefore, likely that there are reversible mechanisms, but this is poorly understood in the pathogenesis of multiple organ dysfunction syndrome (MODS). AIMS Based on our knowledge of extracellular histones and pneumolysin, as endogenous and exogenous pore-forming toxins, respectively, here we clarify if the extent of cell membrane disruption and recovery is important in MODS. METHODS This is a combination of retrospective clinical studies of a cohort of 98 patients from an intensive care unit (ICU) in a tertiary hospital, with interventional animal models and laboratory investigation. RESULTS In patients without septic shock and/or disseminate intravascular coagulation (DIC), circulating histones also strongly correlated with sequential organ failure assessment (SOFA) scores, suggesting their pore-forming property might play an important role. In vivo, histones or pneumolysin infusion similarly caused significant elevation of cell damage markers and multiple organ injury. In trauma and sepsis models, circulating histones strongly correlated with these markers, and anti-histone reagents significantly reduced their release. Comparison of pneumolysin deletion and its parental strain-induced sepsis mouse model showed that pneumolysin was not essential for sepsis development, but enhanced multiple organ damage and reduced survival time. In vitro, histones and pneumolysin treatment disrupt cell membrane integrity, resulting in changes in whole-cell currents and elevated intracellular Ca2+ to lead to Ca2+ overload. Cell-specific damage markers, lactate dehydrogenase (LDH), alanine aminotransferase (ALT), and cardiac troponin I (cTnI), were released from damaged cells. Once toxins were removed, cell membrane damage could be rapidly repaired and cellular function recovered. CONCLUSION This work has confirmed the importance of pore-forming toxins in the development of MODS and proposed a potential mechanism to explain the reversibility of MODS. This may form the foundation for the development of effective therapies.
Collapse
|
15
|
Nishimura N, Fukuda H. Risk of cardiovascular events leading to hospitalisation after Streptococcus pneumoniae infection: a retrospective cohort LIFE Study. BMJ Open 2022; 12:e059713. [PMID: 36332949 PMCID: PMC9639073 DOI: 10.1136/bmjopen-2021-059713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVES To elucidate the risk of cardiovascular event occurrence following Streptococcus pneumoniae infection. DESIGN Retrospective cohort study using a LIFE Study database. SETTING Three municipalities in Japan. PARTICIPANTS Municipality residents who were enrolled in either National Health Insurance or the Latter-Stage Elderly Healthcare System from April 2014 to March 2020. EXPOSURE Occurrence of S. pneumoniae infection. PRIMARY OUTCOME MEASURES Occurrence of one of the following cardiovascular events that led to hospitalisation after S. pneumoniae infection: (1) coronary heart disease (CHD), (2) heart failure (HF), (3) stroke or (4) atrial fibrillation (AF). RESULTS S. pneumoniae-infected patients were matched with non-infected patients for each cardiovascular event. We matched 209 infected patients and 43 499 non-infected patients for CHD, 179 infected patients and 44 148 non-infected patients for HF, 221 infected patients and 44 768 non-infected patients for stroke, and 241 infected patients and 39 568 non-infected patients for AF. During follow-up, the incidence rates for the matched infected and non-infected patients were, respectively, 38.6 (95% CI 19.9 to 67.3) and 30.4 (29.1 to 31.8) per 1000 person-years for CHD; 69.6 (41.9 to 108.8) and 50.5 (48.9 to 52.2) per 1000 person-years for HF; 75.4 (48.3 to 112.2) and 35.5 (34.1 to 36.9) per 1000 person-years for stroke; and 34.7 (17.9 to 60.6) and 11.2 (10.4 to 12.0) per 1000 person-years for AF. Infected patients were significantly more likely to develop stroke (adjusted HR: 2.05, 95% CI 1.22 to 3.47; adjusted subdistribution HR: 1.94, 95% CI 1.15 to 3.26) and AF (3.29, 1.49 to 7.26; 2.74, 1.24 to 6.05) than their non-infected counterparts. CONCLUSIONS S. pneumoniae infections elevate the risk of subsequent stroke and AF occurrence. These findings indicate that pneumococcal infections have short-term effects on patients' health and increase their midterm to long-term susceptibility to serious cardiovascular events.
Collapse
Affiliation(s)
- Naoaki Nishimura
- Department of Medicine, Kyushu University School of Medicine, Fukuoka, Japan
| | - Haruhisa Fukuda
- Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
16
|
de-Miguel-Yanes JM, Jiménez-García R, Hernández-Barrera V, de-Miguel-Díez J, Méndez-Bailón M, López-de-Andrés A. Analysis of the association between atrial fibrillation with in-hospital mortality in people admitted for community-acquired pneumonia through an observational, nation-wide, sex-stratified study. Sci Rep 2022; 12:14404. [PMID: 36002582 PMCID: PMC9402531 DOI: 10.1038/s41598-022-18810-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022] Open
Abstract
We aimed to analyze the influence of atrial fibrillation (AF) prior to hospital admission (“prevalent”) and AF diagnosed during hospital admission (“incident”) on in-hospital mortality (IHM) in women and men admitted for community-acquired pneumonia (CAP) in Spain (2016–2019). We used the Spanish Register of Specialized Care‐Basic Minimum Database. We analyzed 519,750 cases of CAP in people ≥ 18 years (213,631 women (41.1%)), out of which people with prevalent AF represented 23.75% (N = 123,440), whereas people with incident AF constituted 0.60% (N = 3154). Versus no AF, crude IHM was significantly higher for prevalent AF (15.24% vs. 11.40%, p < 0.001) and for incident AF (23.84% vs. 12.24%, p < 0.001). After propensity score marching, IHM in women and men with prevalent AF neared IHM in women and men with no AF (15.72% vs. 15.52%, p = 0.425; and 14.90% vs. 14.99%, p = 0.631, respectively), but IHM in women and men with incident AF was higher than IHM in women and men with no AF (24.37% vs. 13.36%, p < 0.001; and 23.94% vs. 14.04%, p < 0.001, respectively). Male sex was associated with a higher IHM in people with prevalent AF (OR 1.06; 95% CI 1.02–1–10), but not in people with incident AF (OR 0.93; 95% CI 0.77–1–13). AF diagnosed during hospital admission was associated with a higher IHM, irrespectively of sex.
Collapse
Affiliation(s)
- José M de-Miguel-Yanes
- Internal Medicine Department, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, 46, Doctor Esquerdo, 28007, Madrid, Spain
| | - Rodrigo Jiménez-García
- Department of Public Health and Maternal and Child Health, Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
| | - Valentín Hernández-Barrera
- Preventive Medicine and Public Health Teaching and Research Unit, Department of Medical Specialties and Public Health, Universidad Rey Juan Carlos, Alcorcón, Madrid, Spain
| | - Javier de-Miguel-Díez
- Respiratory Care Department, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Hospital General Universitario Gregorio Marañón, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Méndez-Bailón
- Internal Medicine Department, Hospital Universitario Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana López-de-Andrés
- Department of Public Health and Maternal and Child Health, Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
17
|
Zhang C, Deng Y, Wang X, Shi L, Zhan B, Hou N, Liu S, Bao M, Chi G, Fang T. Alnustone inhibits Streptococcus pneumoniae virulence by targeting pneumolysin and sortase A. Fitoterapia 2022; 162:105261. [DOI: 10.1016/j.fitote.2022.105261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 11/04/2022]
|
18
|
Jaiswal V, Ang SP, Lnu K, Ishak A, Pokhrel NB, Chia JE, Hajra A, Biswas M, Matetic A, Dhatt R, Mamas MA. Effect of Pneumococcal Vaccine on Mortality and Cardiovascular Outcomes: A Systematic Review and Meta-Analysis. J Clin Med 2022; 11:3799. [PMID: 35807082 PMCID: PMC9267914 DOI: 10.3390/jcm11133799] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022] Open
Abstract
Various studies have suggested the possible cardiovascular (CV) protective effects of the pneumococcal vaccine (PV). Therefore, we conducted a meta-analysis to assess the association between recipients of PV with mortality and CV outcomes among patients with and without established cardiovascular disease. We performed a systematic literature search in PubMed, Embase, and Scopus for studies evaluating the effect of PV on mortality and CV outcomes. A total of 15 studies with 347,444 patients were included in the meta-analysis: 111,784 patients received PV (32%) and 235,660 patients were in the unvaccinated group (68%). Recipients of PV were associated with decreased all-cause mortality (HR, 0.76 (95% CI: 0.66 to 0.87), p < 0.001). PV was associated with a decrease in the incidence of myocardial infarction (MI) (HR, 0.73 (95% CI: 0.56−0.96), p = 0.02), without significant reduction in CV mortality (HR, 0.87 (95% CI: 0.72−1.07), p = 0.18) and stroke (HR, 1.01 (95% CI: 0.93−1.10), p = 0.82). Our study found PV was associated with decreased risk of all-cause mortality and MI. Future RCTs will be necessary to confirm benefits associated with receipt of PV.
Collapse
Affiliation(s)
- Vikash Jaiswal
- Department of Medicine, Larkin Community Hospital, South Miami, FL 33143, USA;
| | - Song Peng Ang
- Department of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, NJ 08755, USA;
| | - Kriti Lnu
- Department of Internal Medicine, UPMC Harrisburg, Harrisburg, PA 17105, USA;
| | - Angela Ishak
- School of Medicine, European University Cyprus, Nicosia 2404, Cyprus;
| | | | - Jia Ee Chia
- School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia;
| | - Adrija Hajra
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, The Bronx, NY 10461, USA;
| | - Monodeep Biswas
- Division of Cardiology, Wellspan Cardiology, Lancaster, PA 17602, USA;
| | - Andrija Matetic
- Department of Cardiology, University Hospital of Split, 21000 Split, Croatia;
- Keele Cardiovascular Research Group, Centre for Prognosis Research, Keele University, Keele ST5 5BG, UK
| | - Ravinder Dhatt
- Department of Internal Medicine, UPMC Harrisburg, Harrisburg, PA 17105, USA;
| | - Mamas A. Mamas
- Keele Cardiovascular Research Group, Centre for Prognosis Research, Keele University, Keele ST5 5BG, UK
| |
Collapse
|
19
|
Oh MW, Lella M, Kuo SH, Tal-Gan Y, Lau GW. Pharmacological Evaluation of Synthetic Dominant-Negative Peptides Derived from the Competence-Stimulating Peptide of Streptococcus pneumoniae. ACS Pharmacol Transl Sci 2022; 5:299-305. [PMID: 35592433 PMCID: PMC9112410 DOI: 10.1021/acsptsci.2c00037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Indexed: 11/28/2022]
Abstract
The competence regulon of Streptococcus pneumoniae (pneumococcus) is a quorum-sensing circuitry that regulates the ability of this pathogen to acquire antibiotic resistance or perform serotype switching, leading to vaccine-escape serotypes, via horizontal gene transfer, as well as initiate virulence. Induction of the competence regulon is centered on binding of the competence-stimulating peptide (CSP) to its cognate receptor, ComD. We have recently synthesized multiple dominant-negative peptide analogs capable of inhibiting competence induction and virulence in S. pneumoniae. However, the pharmacodynamics and safety profiles of these peptide drug leads have not been characterized. Therefore, in this study, we compared the biostability of cyanine-7.5-labeled wild-type CSPs versus dominant-negative peptide analogs (dnCSPs) spatiotemporally by using an IVIS Spectrum in vivo imaging system. Moreover, in vitro cytotoxicity and in vivo toxicity were evaluated. We conclude that our best peptide analog, CSP1-E1A-cyc(Dap6E10), is an attractive therapeutic agent against pneumococcal infection with superior safety and pharmacokinetics profiles.
Collapse
Affiliation(s)
- Myung Whan Oh
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61802, United States
| | - Muralikrishna Lella
- Department of Chemistry, University of Nevada, Reno, 1664 North Virginia Street, Reno, Nevada 89557, United States
| | - Shanny Hsuan Kuo
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61802, United States
| | - Yftah Tal-Gan
- Department of Chemistry, University of Nevada, Reno, 1664 North Virginia Street, Reno, Nevada 89557, United States
| | - Gee W Lau
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61802, United States
| |
Collapse
|
20
|
Wang Q, Wang X, Lv Y, Yang C, Zhou C, Wang L. Changes in Rats' Gut Microbiota Composition Caused by Induced Chronic Myocardial Infarction Lead to Depression-Like Behavior. Front Microbiol 2022; 12:641084. [PMID: 35496818 PMCID: PMC9048826 DOI: 10.3389/fmicb.2021.641084] [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: 12/13/2020] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
Depression is common among patients who have chronic myocardial infarction (CMI). Despite their frequency, depression and CMI are bidirectional related conditions, each is a risk for the other, and they often co-exist, suggesting shared or interacting pathomechanisms. Accumulating data revealed the effects of gut microbiota in terms of regulating depression via the gut–brain axis. Thus, we investigated the role of gut microbial dysbiosis in CMI-induced depression-like behavior. Hierarchical cluster analysis of sucrose preference test (SPT) results was adopted to classify the CMI rats into depression-like behavior (CMI + Dep) or non-depression-like behavior (CMI + Non-Dep) phenotypes. First, 16S ribosomal RNA sequencing analysis showed both β-diversity and relative abundance of several gut bacteria significantly differed between the CMI + Dep and CMI + Non-Dep rats. Next, transplantation of fecal microbiota from CMI + Dep rats visibly altered the relative abundance of gut microbiota and also induced depression-like behavior in the antibiotics-treated pseudo-germ-free rats. In conclusion, these findings suggested that dysbiosis in gut microbial composition contributed to the onset of CMI-induced depression-like behavior and that exogenous regulation of gut microbiota composition could be a potential therapeutic strategy for CMI and related depression-like behavior.
Collapse
Affiliation(s)
- Qianwen Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yong Lv
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chun Yang
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chenliang Zhou
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Long Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
21
|
Pereira JM, Xu S, Leong JM, Sousa S. The Yin and Yang of Pneumolysin During Pneumococcal Infection. Front Immunol 2022; 13:878244. [PMID: 35529870 PMCID: PMC9074694 DOI: 10.3389/fimmu.2022.878244] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/23/2022] [Indexed: 12/15/2022] Open
Abstract
Pneumolysin (PLY) is a pore-forming toxin produced by the human pathobiont Streptococcus pneumoniae, the major cause of pneumonia worldwide. PLY, a key pneumococcal virulence factor, can form transmembrane pores in host cells, disrupting plasma membrane integrity and deregulating cellular homeostasis. At lytic concentrations, PLY causes cell death. At sub-lytic concentrations, PLY triggers host cell survival pathways that cooperate to reseal the damaged plasma membrane and restore cell homeostasis. While PLY is generally considered a pivotal factor promoting S. pneumoniae colonization and survival, it is also a powerful trigger of the innate and adaptive host immune response against bacterial infection. The dichotomy of PLY as both a key bacterial virulence factor and a trigger for host immune modulation allows the toxin to display both "Yin" and "Yang" properties during infection, promoting disease by membrane perforation and activating inflammatory pathways, while also mitigating damage by triggering host cell repair and initiating anti-inflammatory responses. Due to its cytolytic activity and diverse immunomodulatory properties, PLY is integral to every stage of S. pneumoniae pathogenesis and may tip the balance towards either the pathogen or the host depending on the context of infection.
Collapse
Affiliation(s)
- Joana M. Pereira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Molecular and Cellular (MC) Biology PhD Program, ICBAS - Instituto de Ciência Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Shuying Xu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, United States
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA, United States
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, United States
| | - Sandra Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
22
|
Kopenhagen A, Ramming I, Camp B, Hammerschmidt S, Fulde M, Müsken M, Steinert M, Bergmann S. Streptococcus pneumoniae Affects Endothelial Cell Migration in Microfluidic Circulation. Front Microbiol 2022; 13:852036. [PMID: 35401456 PMCID: PMC8990767 DOI: 10.3389/fmicb.2022.852036] [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: 01/10/2022] [Accepted: 02/04/2022] [Indexed: 01/12/2023] Open
Abstract
Bloodstream infections caused by Streptococcus pneumoniae induce strong inflammatory and procoagulant cellular responses and affect the endothelial barrier of the vascular system. Bacterial virulence determinants, such as the cytotoxic pore-forming pneumolysin, increase the endothelial barrier permeability by inducing cell apoptosis and cell damage. As life-threatening consequences, disseminated intravascular coagulation followed by consumption coagulopathy and low blood pressure is described. With the aim to decipher the role of pneumolysin in endothelial damage and leakage of the vascular barrier in more detail, we established a chamber-separation cell migration assay (CSMA) used to illustrate endothelial wound healing upon bacterial infections. We used chambered inlets for cell cultivation, which, after removal, provide a cell-free area of 500 μm in diameter as a defined gap in primary endothelial cell layers. During the process of wound healing, the size of the cell-free area is decreasing due to cell migration and proliferation, which we quantitatively determined by microscopic live cell monitoring. In addition, differential immunofluorescence staining combined with confocal microscopy was used to morphologically characterize the effect of bacterial attachment on cell migration and the velocity of gap closure. In all assays, the presence of wild-type pneumococci significantly inhibited endothelial gap closure. Remarkably, even in the presence of pneumolysin-deficient pneumococci, cell migration was significantly retarded. Moreover, the inhibitory effect of pneumococci on the proportion of cell proliferation versus cell migration within the process of endothelial gap closure was assessed by implementation of a fluorescence-conjugated nucleoside analogon. We further combined the endothelial CSMA with a microfluidic pump system, which for the first time enabled the microscopic visualization and monitoring of endothelial gap closure in the presence of circulating bacteria at defined vascular shear stress values for up to 48 h. In accordance with our CSMA results under static conditions, the gap remained cell free in the presence of circulating pneumococci in flow. Hence, our combined endothelial cultivation technique represents a complex in vitro system, which mimics the vascular physiology as close as possible by providing essential parameters of the blood flow to gain new insights into the effect of pneumococcal infection on endothelial barrier integrity in flow.
Collapse
Affiliation(s)
- Anna Kopenhagen
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany
| | - Isabell Ramming
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Infectious Diseases, Robert Koch Institute, Wernigerode, Germany
| | - Belinda Camp
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany.,Department of Pneumology, University Hospital Magdeburg, Magdeburg, Germany
| | - Sven Hammerschmidt
- Institute for Genetics and Functional Genomics, Department of Molecular Genetics and Infection Biology, Universität Greifswald, Greifswald, Germany
| | - Marcus Fulde
- Institute of Microbiology and Epizootics, Department of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Steinert
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany.,Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Simone Bergmann
- Institut für Mikrobiologie, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
23
|
Lata K, Singh M, Chatterjee S, Chattopadhyay K. Membrane Dynamics and Remodelling in Response to the Action of the Membrane-Damaging Pore-Forming Toxins. J Membr Biol 2022; 255:161-173. [PMID: 35305136 DOI: 10.1007/s00232-022-00227-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/02/2022] [Indexed: 12/11/2022]
Abstract
Pore-forming protein toxins (PFTs) represent a diverse class of membrane-damaging proteins that are produced by a wide variety of organisms. PFT-mediated membrane perforation is largely governed by the chemical composition and the physical properties of the plasma membranes. The interaction between the PFTs with the target membranes is critical for the initiation of the pore-formation process, and can lead to discrete membrane reorganization events that further aids in the process of pore-formation. Punching holes on the plasma membranes by the PFTs interferes with the cellular homeostasis by disrupting the ion-balance inside the cells that in turn can turn on multiple signalling cascades required to restore membrane integrity and cellular homeostasis. In this review, we discuss the physicochemical attributes of the plasma membranes associated with the pore-formation processes by the PFTs, and the subsequent membrane remodelling events that may start off the membrane-repair mechanisms.
Collapse
Affiliation(s)
- Kusum Lata
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India
| | - Mahendra Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India
| | - Shamaita Chatterjee
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India
| | - Kausik Chattopadhyay
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar, Manauli, Mohali, Punjab, 140306, India.
| |
Collapse
|
24
|
The Influence of Atrial Fibrillation on In-Hospital Mortality in People with Hospital-Acquired Pneumonia: An Observational, Sex-Stratified Study. J Clin Med 2022; 11:jcm11051179. [PMID: 35268270 PMCID: PMC8910951 DOI: 10.3390/jcm11051179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/16/2022] [Accepted: 02/21/2022] [Indexed: 12/23/2022] Open
Abstract
(1) Background: The study aimed to analyze the influence of atrial fibrillation (AF) prior to hospital admission (“prevalent”) and new-onset AF diagnosed during hospital admission (“incident”) on in-hospital mortality (IHM) in women and men who developed hospital-acquired pneumonia (HAP) in Spain (2016−2019). (2) Methods: We used the Spanish Register of Specialized Care-Basic Minimum Database. (3) Results: We analyzed 38,814 cases of HAP (34.6% women; 13.5% ventilator-associated). Prevalent AF was coded in 19.9% (n = 7742), and incident AF in 5.5% (n = 2136) of HAP. Crude IHM was significantly higher for prevalent AF (34.22% vs. 27.35%, p < 0.001) and for incident AF (35.81% vs. 28.31%, p < 0.001) compared to no AF. After propensity score matching, IHM among women and men with prevalent AF was higher than among women and men with no AF (among women, 32.89% vs. 30.11%, p = 0.021; among men, 35.05% vs. 32.46%, p = 0.008). Similarly, IHM among women and men with incident AF was higher than among women and men with no AF (among women, 36.23% vs. 29.90%, p = 0.013; among men, 35.62% vs. 30.47%; p = 0.003). Sex was associated with a higher IHM only in people with incident AF (for female, OR = 1.21; 95% CI: 1.01−1.57). (4) Conclusions: Both prevalent and incident AF were associated with higher IHM in people who developed HAP. Female sex was associated with a higher IHM in incident AF.
Collapse
|
25
|
Klugman KP, Feldman C. Serotype-specific Cardiac Involvement in Pneumococcal Pneumonia. Clin Infect Dis 2022; 74:507-508. [PMID: 32964220 PMCID: PMC8834653 DOI: 10.1093/cid/ciaa1434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Keith P Klugman
- Department of Global Health, School of Public Health, Emory University, Atlanta, Georgia, USA.,Department of Medical Microbiology, School of Pathology, University of the Witwatersrand Medical School, Johannesburg, South Africa.,Pneumonia Program, Bill and Melinda Gates Foundation, Seattle, Washington, USA
| | - Charles Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
26
|
Sasagawa K, Domon H, Sakagami R, Hirayama S, Maekawa T, Isono T, Hiyoshi T, Tamura H, Takizawa F, Fukushima Y, Tabeta K, Terao Y. Matcha Green Tea Exhibits Bactericidal Activity against Streptococcus pneumoniae and Inhibits Functional Pneumolysin. Antibiotics (Basel) 2021; 10:antibiotics10121550. [PMID: 34943762 PMCID: PMC8698834 DOI: 10.3390/antibiotics10121550] [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: 11/17/2021] [Revised: 12/07/2021] [Accepted: 12/16/2021] [Indexed: 11/16/2022] Open
Abstract
Streptococcus pneumoniae is a causative pathogen of several human infectious diseases including community-acquired pneumonia. Pneumolysin (PLY), a pore-forming toxin, plays an important role in the pathogenesis of pneumococcal pneumonia. In recent years, the use of traditional natural substances for prevention has drawn attention because of the increasing antibacterial drug resistance of S. pneumoniae. According to some studies, green tea exhibits antibacterial and antitoxin activities. The polyphenols, namely the catechins epigallocatechin gallate (EGCG), epigallocatechin (EGC), epicatechin gallate (ECG), and epicatechin (EC) are largely responsible for these activities. Although matcha green tea provides more polyphenols than green tea infusions, its relationship with pneumococcal pneumonia remains unclear. In this study, we found that treatment with 20 mg/mL matcha supernatant exhibited significant antibacterial activity against S. pneumoniae regardless of antimicrobial resistance. In addition, the matcha supernatant suppressed PLY-mediated hemolysis and cytolysis by inhibiting PLY oligomerization. Moreover, the matcha supernatant and catechins inhibited PLY-mediated neutrophil death and the release of neutrophil elastase. These findings suggest that matcha green tea reduces the virulence of S. pneumoniae in vitro and may be a promising agent for the treatment of pneumococcal infections.
Collapse
Affiliation(s)
- Karin Sasagawa
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| | - Hisanori Domon
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Rina Sakagami
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
| | - Satoru Hirayama
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
| | - Tomoki Maekawa
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Toshihito Isono
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
| | - Takumi Hiyoshi
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
| | - Hikaru Tamura
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| | - Fumio Takizawa
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| | - Yoichi Fukushima
- Nestlé Japan Ltd., Wellness Communications, Tokyo 140-0002, Japan;
| | - Koichi Tabeta
- Division of Periodontology, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan;
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8514, Japan; (K.S.); (H.D.); (R.S.); (S.H.); (T.M.); (T.I.); (T.H.); (H.T.); (F.T.)
- Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8514, Japan
- Correspondence: ; Tel.: +81-25-227-2838
| |
Collapse
|
27
|
Categorizing sequences of concern by function to better assess mechanisms of microbial pathogenesis. Infect Immun 2021; 90:e0033421. [PMID: 34780277 PMCID: PMC9119117 DOI: 10.1128/iai.00334-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
To identify sequences with a role in microbial pathogenesis, we assessed the adequacy of their annotation by existing controlled vocabularies and sequence databases. Our goal was to regularize descriptions of microbial pathogenesis for improved integration with bioinformatic applications. Here, we review the challenges of annotating sequences for pathogenic activity. We relate the categorization of more than 2,750 sequences of pathogenic microbes through a controlled vocabulary called Functions of Sequences of Concern (FunSoCs). These allow for an ease of description by both humans and machines. We provide a subset of 220 fully annotated sequences in the supplemental material as examples. The use of this compact (∼30 terms), controlled vocabulary has potential benefits for research in microbial genomics, public health, biosecurity, biosurveillance, and the characterization of new and emerging pathogens.
Collapse
|
28
|
Streptolysin O concentration and activity is central to in vivo phenotype and disease outcome in Group A Streptococcus infection. Sci Rep 2021; 11:19011. [PMID: 34561464 PMCID: PMC8463576 DOI: 10.1038/s41598-021-97866-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 08/23/2021] [Indexed: 11/08/2022] Open
Abstract
Group A Streptoccocus (GAS) is among the most diverse of all human pathogens, responsible for a range of clinical manifestations, from mild superficial infections such as pharyngitis to serious invasive infections such as necrotising fasciitis and sepsis. The drivers of these different disease phenotypes are not known. The GAS cholesterol-dependent cytolysin, Streptolysin O (SLO), has well established cell and tissue destructive activity. We investigated the role of SLO in determining disease outcome in vivo, by using two different clinical lineages; the recently emerged hypervirulent outbreak emm type 32.2 strains, which result in sepsis, and the emm type 1.0 strains which cause septic arthritis. Using clinically relevant in vivo mouse models of sepsis and a novel septic arthritis model, we found that the amount and activity of SLO was vital in determining the course of infection. The emm type 32.2 strain produced large quantities of highly haemolytic SLO that resulted in rapid development of sepsis. By contrast, the reduced concentration and lower haemolytic activity of emm type 1.0 SLO led to translocation of bacteria from blood to joints. Importantly, sepsis associated strains that were attenuated by deletion or inhibition of SLO, then also translocated to the joint, confirming the key role of SLO in determining infection niche. Our findings demonstrate that SLO is key to in vivo phenotype and disease outcome. Careful consideration should be given to novel therapy or vaccination strategies that target SLO. Whilst neutralising SLO activity may reduce severe invasive disease, it has the potential to promote chronic inflammatory conditions such as septic arthritis.
Collapse
|
29
|
Pneumolysin induces platelet destruction, not platelet activation, which can be prevented by immunoglobulin preparations in vitro. Blood Adv 2021; 4:6315-6326. [PMID: 33351126 DOI: 10.1182/bloodadvances.2020002372] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 11/05/2020] [Indexed: 01/08/2023] Open
Abstract
Community-acquired pneumonia by primary or superinfections with Streptococcus pneumoniae can lead to acute respiratory distress requiring mechanical ventilation. The pore-forming toxin pneumolysin alters the alveolar-capillary barrier and causes extravasation of protein-rich fluid into the interstitial pulmonary tissue, which impairs gas exchange. Platelets usually prevent endothelial leakage in inflamed pulmonary tissue by sealing inflammation-induced endothelial gaps. We not only confirm that S pneumoniae induces CD62P expression in platelets, but we also show that, in the presence of pneumolysin, CD62P expression is not associated with platelet activation. Pneumolysin induces pores in the platelet membrane, which allow anti-CD62P antibodies to stain the intracellular CD62P without platelet activation. Pneumolysin treatment also results in calcium efflux, increase in light transmission by platelet lysis (not aggregation), loss of platelet thrombus formation in the flow chamber, and loss of pore-sealing capacity of platelets in the Boyden chamber. Specific anti-pneumolysin monoclonal and polyclonal antibodies inhibit these effects of pneumolysin on platelets as do polyvalent human immunoglobulins. In a post hoc analysis of the prospective randomized phase 2 CIGMA trial, we show that administration of a polyvalent immunoglobulin preparation was associated with a nominally higher platelet count and nominally improved survival in patients with severe S pneumoniae-related community-acquired pneumonia. Although, due to the low number of patients, no definitive conclusion can be made, our findings provide a rationale for investigation of pharmacologic immunoglobulin preparations to target pneumolysin by polyvalent immunoglobulin preparations in severe community-acquired pneumococcal pneumonia, to counteract the risk of these patients becoming ventilation dependent. This trial was registered at www.clinicaltrials.gov as #NCT01420744.
Collapse
|
30
|
Scott NR, Mann B, Tuomanen EI, Orihuela CJ. Multi-Valent Protein Hybrid Pneumococcal Vaccines: A Strategy for the Next Generation of Vaccines. Vaccines (Basel) 2021; 9:209. [PMID: 33801372 PMCID: PMC8002124 DOI: 10.3390/vaccines9030209] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/15/2022] Open
Abstract
Streptococcus pneumoniae (Spn) is a bacterial pathogen known to colonize the upper respiratory tract and cause serious opportunistic diseases such as pneumonia, bacteremia, sepsis and meningitis. As a consequence, millions of attributable deaths occur annually, especially among infants, the elderly and immunocompromised individuals. Although current vaccines, composed of purified pneumococcal polysaccharide in free form or conjugated to a protein carrier, are widely used and have been demonstrated to be effective in target groups, Spn has continued to colonize and cause life-threatening disease in susceptible populations. This lack of broad protection highlights the necessity of improving upon the current "gold standard" pneumococcal vaccines to increase protection both by decreasing colonization and reducing the incidence of sterile-site infections. Over the past century, most of the pneumococcal proteins that play an essential role in colonization and pathogenesis have been identified and characterized. Some of these proteins have the potential to serve as antigens in a multi-valent protein vaccine that confers capsule independent protection. This review seeks to summarize the benefits and limitations of the currently employed vaccine strategies, describes how leading candidate proteins contribute to pneumococcal disease development, and discusses the potential of these proteins as protective antigens-including as a hybrid construct.
Collapse
Affiliation(s)
- Ninecia R. Scott
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Beth Mann
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (B.M.); (E.I.T.)
| | - Elaine I. Tuomanen
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (B.M.); (E.I.T.)
| | - Carlos J. Orihuela
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| |
Collapse
|
31
|
Algowhary M. Association between age and infection in patients with acute ST-elevation myocardial infarction. Egypt Heart J 2021; 73:12. [PMID: 33515355 PMCID: PMC7847417 DOI: 10.1186/s43044-021-00137-w] [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: 10/10/2020] [Accepted: 01/21/2021] [Indexed: 12/13/2022] Open
Abstract
Background ST-elevation myocardial infarction (STEMI) in young patients has a unique risk profile. We aimed to detect bacteria in aspirate of infarct artery in young versus old patients. Results Aspirates of consecutive 140 patients who underwent a primary coronary intervention were taken for bacteriological, microscopical, and immunohistochemical (for bacterial pneumolysin) examinations. Their results were calculated in young (≤ 50 years) versus old (> 50 years) patients. Median age (interquartile range) was 45 (38–48) years in young (60 patients) and 59 (55–65) years in old (80 patients) patients, p < 0.0001. Both groups had similar baseline data except age, males, diabetes, hyperlipidemia, family history, lesion length, and ectatic vessel. Different bacteria were cultured in 11.3% of all patients involving 22.6% of young and 2.8% of old patients [hazard ratio 8.03 (95% CI 1.83–51.49), p = 0.002]. By multivariate analyses, age groups and leukocytic count were independent predictors of infection (bacteria and pneumolysin), p = 0.027 and p < 0.0001, respectively. Optimal cutoff value of leukocytic count was 12,250 cells/μl [ROC curve sensitivity 85.7%, specificity 86.4%, and AUC 0.97 (95% CI 0.95–1.0), p < 0.001]. Infection was an independent predictor of STEMI in young versus old patients, p < 0.001. Nevertheless, in-hospital events occurred insignificantly different and neither age groups nor infection was predictor of in-hospital events. Conclusions Young patients had significantly higher percentage of bacteria in their infarcted artery than old patients. High leukocytic count in patients below 50 predicts infection that causes acute myocardial infarction. Antibacterial trials directed toward this group are required for secondary prevention.
Collapse
Affiliation(s)
- Magdy Algowhary
- Department of Cardiovascular Medicine, Assiut University Heart Hospital, Assiut University, Asyut, 71515, Egypt.
| |
Collapse
|
32
|
Pore-forming toxins in infection and immunity. Biochem Soc Trans 2021; 49:455-465. [PMID: 33492383 DOI: 10.1042/bst20200836] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/02/2021] [Accepted: 01/06/2021] [Indexed: 12/14/2022]
Abstract
The integrity of the plasma membranes is extremely crucial for the survival and proper functioning of the cells. Organisms from all kingdoms of life employ specialized pore-forming proteins and toxins (PFPs and PFTs) that perforate cell membranes, and cause detrimental effects. PFPs/PFTs exert their damaging actions by forming oligomeric pores in the membrane lipid bilayer. PFPs/PFTs play important roles in diverse biological processes. Many pathogenic bacteria secrete PFTs for executing their virulence mechanisms. The immune system of the higher vertebrates employs PFPs to kill pathogen-infected cells and transformed cancer cells. The most obvious consequence of membrane pore-formation by the PFPs/PFTs is the killing of the target cells due to the disruption of the permeability barrier function of the plasma membranes. PFPs/PFTs can also activate diverse cellular processes that include activation of the stress-response pathways, induction of programmed cell death, and inflammation. Upon attack by the PFTs, host cells may also activate pathways to repair the injured membranes, restore cellular homeostasis, and trigger inflammatory immune responses. In this article, we present an overview of the diverse cellular responses that are triggered by the PFPs/PFTs, and their implications in the process of pathogen infection and immunity.
Collapse
|
33
|
Lafuse WP, Wozniak DJ, Rajaram MVS. Role of Cardiac Macrophages on Cardiac Inflammation, Fibrosis and Tissue Repair. Cells 2020; 10:E51. [PMID: 33396359 PMCID: PMC7824389 DOI: 10.3390/cells10010051] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022] Open
Abstract
The immune system plays a pivotal role in the initiation, development and resolution of inflammation following insult or damage to organs. The heart is a vital organ which supplies nutrients and oxygen to all parts of the body. Heart failure (HF) has been conventionally described as a disease associated with cardiac tissue damage caused by systemic inflammation, arrhythmia and conduction defects. Cardiac inflammation and subsequent tissue damage is orchestrated by the infiltration and activation of various immune cells including neutrophils, monocytes, macrophages, eosinophils, mast cells, natural killer cells, and T and B cells into the myocardium. After tissue injury, monocytes and tissue-resident macrophages undergo marked phenotypic and functional changes, and function as key regulators of tissue repair, regeneration and fibrosis. Disturbance in resident macrophage functions such as uncontrolled production of inflammatory cytokines, growth factors and inefficient generation of an anti-inflammatory response or unsuccessful communication between macrophages and epithelial and endothelial cells and fibroblasts can lead to aberrant repair, persistent injury, and HF. Therefore, in this review, we discuss the role of cardiac macrophages on cardiac inflammation, tissue repair, regeneration and fibrosis.
Collapse
Affiliation(s)
- William P. Lafuse
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (W.P.L.); (D.J.W.)
| | - Daniel J. Wozniak
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (W.P.L.); (D.J.W.)
- Department of Microbiology, Ohio State University, Columbus, OH 43210, USA
| | - Murugesan V. S. Rajaram
- Department of Microbial Infection and Immunity, College of Medicine, Ohio State University, Columbus, OH 43210, USA; (W.P.L.); (D.J.W.)
| |
Collapse
|
34
|
Tonon F, Di Bella S, Grassi G, Luzzati R, Ascenzi P, di Masi A, Zennaro C. Extra-Intestinal Effects of C.difficile Toxin A and B: An In Vivo Study Using the Zebrafish Embryo Model. Cells 2020; 9:2575. [PMID: 33271969 PMCID: PMC7760802 DOI: 10.3390/cells9122575] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/18/2022] Open
Abstract
C.difficile infection (CDI) is not a merely "gut-confined" disease as toxemia could drive the development of CDI-related extra-intestinal effects. These effects could explain the high CDI-associated mortality, not just justified by diarrhea and dehydration. Here, the extra-intestinal effects of toxin A (TcdA) and B (TcdB) produced by C. difficile have been studied in vivo using the zebrafish embryo model. Noteworthy, protective properties of human serum albumin (HSA) towards toxins-induced extra-intestinal effects were also addressed. Zebrafish embryos were treated with TcdA, TcdB and/or HSA at 24 h post-fertilization. Embryos were analyzed for 48 h after treatment to check vital signs and morphological changes. Markers related to cardio-vascular damage and inflammation were evaluated by Real-Time quantitative PCR and/or western blotting. Both toxins induced cardiovascular damage in zebrafish embryos by different mechanisms: (i) direct toxicity (i.e., pericardial edema, cardiac chambers enlargement, endothelial alteration); (ii) increased hormonal production and release (i.e., atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP)), (iii) alteration of the vascular system through the increase of the vascular endothelial growth factor (VEGF-A) levels, as well as of its receptors, (iv) pro-inflammatory response through high cytokines production (i.e., CXCL8, IL1B, IL6 and TNFα) and (v) cell-mediated damage due to the increase in neutrophils number. In addition to cardiovascular damage, we observe skin alteration and inflammation. Finally, our data indicate a protective effect of HSA toward the toxins induced extra-intestinal effects. Together, our findings can serve as a starting point for humans' studies to substantiate and understand the extra-intestinal effects observed in CDI patients.
Collapse
Affiliation(s)
- Federica Tonon
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (S.D.B.); (R.L.); (C.Z.)
| | - Stefano Di Bella
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (S.D.B.); (R.L.); (C.Z.)
| | - Gabriele Grassi
- Department of Life Sciences, University of Trieste, 34128 Trieste, Italy;
| | - Roberto Luzzati
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (S.D.B.); (R.L.); (C.Z.)
| | - Paolo Ascenzi
- Department of Sciences, Roma Tre University, 00154 Rome, Italy; (P.A.); (A.d.M.)
| | - Alessandra di Masi
- Department of Sciences, Roma Tre University, 00154 Rome, Italy; (P.A.); (A.d.M.)
| | - Cristina Zennaro
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34149 Trieste, Italy; (S.D.B.); (R.L.); (C.Z.)
| |
Collapse
|
35
|
Rombauts A, Abelenda-Alonso G, Càmara J, Lorenzo-Esteller L, González-Díaz A, Sastre-Escolà E, Gudiol C, Dorca J, Tebé C, Pallarès N, Ardanuy C, Carratalà J. Host- and Pathogen-Related Factors for Acute Cardiac Events in Pneumococcal Pneumonia. Open Forum Infect Dis 2020; 7:ofaa522. [PMID: 33335932 PMCID: PMC7727332 DOI: 10.1093/ofid/ofaa522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
Background Acute cardiac events (ACEs) are increasingly being recognized as a major complication in pneumococcal community-acquired pneumonia (CAP). Information regarding host- and pathogen-related factors for ACEs, including pneumococcal serotypes and clonal complexes, is scarce. Methods A retrospective study was conducted of a prospective cohort of patients hospitalized for CAP between 1996 and 2019. Logistic regression and funnel plot analyses were performed to determine host- and pathogen-related factors for ACEs. Results Of 1739 episodes of pneumococcal CAP, 1 or more ACEs occurred in 304 (17.5%) patients, the most frequent being arrhythmia (n = 207), heart failure (n = 135), and myocardial infarction (n = 23). The majority of ACEs (73.4%) occurred within 48 hours of admission. Factors independently associated with ACEs were older age, preexisting heart conditions, pneumococcal bacteremia, septic shock at admission, and high-risk pneumonia. Among 983 pneumococcal isolates, 872 (88.7%) were serotyped and 742 (75.5%) genotyped. The funnel plot analyses did not find any statistically significant association between serotypes or clonal complexes with ACEs. Nevertheless, there was a trend toward an association between CC230 and these complications. ACEs were independently associated with 30-day mortality (adjusted odds ratio, 1.88; 95% CI, 1.11-3.13). Conclusions ACEs are frequent in pneumococcal pneumonia and are associated with increased mortality. The risk factors defined in this study may help identify patients who must undergo close follow-up, including heart rhythm monitoring, and special care to avoid fluid overload, particularly during the first 48 hours of admission. These high-risk patients should be the target for preventive intervention strategies.
Collapse
Affiliation(s)
- Alexander Rombauts
- Department of Infectious Diseases, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Gabriela Abelenda-Alonso
- Department of Infectious Diseases, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Càmara
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.,Department of Microbiology, Bellvitge Universtiy Hospital, Hospitalet de Llobregat, Barcelona, Spain.,Research Network for Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Laia Lorenzo-Esteller
- Department of Infectious Diseases, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
| | - Aida González-Díaz
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.,Department of Microbiology, Bellvitge Universtiy Hospital, Hospitalet de Llobregat, Barcelona, Spain.,Research Network for Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Enric Sastre-Escolà
- Department of Infectious Diseases, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
| | - Carlota Gudiol
- Department of Infectious Diseases, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.,Spanish Network for Research in Infectious Diseases (REIPI), Instituto de Salud Carlos III, Madrid, Spain.,University of Barcelona, Barcelona, Spain
| | - Jordi Dorca
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.,University of Barcelona, Barcelona, Spain.,Department of Pneumology, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
| | - Cristian Tebé
- Biostatistics Unit at Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Natàlia Pallarès
- Biostatistics Unit at Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Carmen Ardanuy
- Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.,Department of Microbiology, Bellvitge Universtiy Hospital, Hospitalet de Llobregat, Barcelona, Spain.,Research Network for Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,University of Barcelona, Barcelona, Spain
| | - Jordi Carratalà
- Department of Infectious Diseases, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain.,Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.,Spanish Network for Research in Infectious Diseases (REIPI), Instituto de Salud Carlos III, Madrid, Spain.,University of Barcelona, Barcelona, Spain
| |
Collapse
|
36
|
Panagiotou S, Chaguza C, Yahya R, Audshasai T, Baltazar M, Ressel L, Khandaker S, Alsahag M, Mitchell TJ, Prudhomme M, Kadioglu A, Yang M. Hypervirulent pneumococcal serotype 1 harbours two pneumolysin variants with differential haemolytic activity. Sci Rep 2020; 10:17313. [PMID: 33057054 PMCID: PMC7560715 DOI: 10.1038/s41598-020-73454-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022] Open
Abstract
Streptococcus pneumoniae is a devastating global pathogen. Prevalent in sub-Saharan Africa, pneumococcal serotype 1 is atypical in that it is rarely found as a nasopharyngeal coloniser, yet is described as one of the most common causes of invasive pneumococcal disease. Clonal sequence type (ST)-306 and ST615 are representative of the two major serotype 1 lineages A and C, respectively. Here we investigated the virulence properties and haemolytic activities of these 2 clonal types using in vivo mouse models and in vitro assays. A lethal dose of ST615 administered intranasally to mice led to the rapid onset of disease symptoms and resulted in 90% mortality. In contrast, mice exposed to the same infection dose of ST306 or a pneumolysin (Ply)-deficient ST615 failed to develop any disease symptoms. Interestingly, the 2 strains did not differ in their ability to bind the immune complement or to undergo neutrophil-mediated phagocytosis. Upon comparative genomic analysis, we found higher within-ST sequence diversity in ST615 compared with ST306 and determined that ZmpA, ZmpD proteins, and IgA protease, were uniquely found in ST615. Using cell fractionation and cell contact-dependent assay, we made the unexpected finding that ST615 harbours the expression of two haemolytic variants of Ply: a cell-wall restricted fully haemolytic Ply, and a cytosolic pool of Ply void of any detectable haemolytic activity. This is the first time such a phenomenon has been described. We discuss the biological significance of our observation in relation to the aptitude of the pneumococcus for sustaining its human reservoir.
Collapse
Affiliation(s)
- Stavros Panagiotou
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK
| | - Chrispin Chaguza
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
- Darwin College, University of Cambridge, Silver Street, Cambridge, CB3 9EU, UK
| | - Reham Yahya
- College of sciences and health professions, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Teerawit Audshasai
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK
| | - Murielle Baltazar
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK
| | - Lorenzo Ressel
- Department of Veterinary Pathology and Public Health, Institute of Veterinary Science, University of Liverpool, Leahurst Campus, Neston, CH64 7TE, UK
| | - Shadia Khandaker
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK
| | - Mansoor Alsahag
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK
- Faculty of Applied Medical Sciences, Albaha University, Albaha, Kingdom of Saudi Arabia
| | - Tim J Mitchell
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Marc Prudhomme
- Université Paul Sabatier, Centre National de la Recherche Scientifique, 118 Route de Narbonne, 31062, Toulouse Cedex 9, France
| | - Aras Kadioglu
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK.
| | - Marie Yang
- Department of Clinical Infection Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, The Ronald Ross Building, 8 West Derby St, Liverpool, L69 7BE, UK.
| |
Collapse
|
37
|
Chaguza C, Yang M, Cornick JE, du Plessis M, Gladstone RA, Kwambana-Adams BA, Lo SW, Ebruke C, Tonkin-Hill G, Peno C, Senghore M, Obaro SK, Ousmane S, Pluschke G, Collard JM, Sigaùque B, French N, Klugman KP, Heyderman RS, McGee L, Antonio M, Breiman RF, von Gottberg A, Everett DB, Kadioglu A, Bentley SD. Bacterial genome-wide association study of hyper-virulent pneumococcal serotype 1 identifies genetic variation associated with neurotropism. Commun Biol 2020; 3:559. [PMID: 33033372 PMCID: PMC7545184 DOI: 10.1038/s42003-020-01290-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/11/2020] [Indexed: 12/19/2022] Open
Abstract
Hyper-virulent Streptococcus pneumoniae serotype 1 strains are endemic in Sub-Saharan Africa and frequently cause lethal meningitis outbreaks. It remains unknown whether genetic variation in serotype 1 strains modulates tropism into cerebrospinal fluid to cause central nervous system (CNS) infections, particularly meningitis. Here, we address this question through a large-scale linear mixed model genome-wide association study of 909 African pneumococcal serotype 1 isolates collected from CNS and non-CNS human samples. By controlling for host age, geography, and strain population structure, we identify genome-wide statistically significant genotype-phenotype associations in surface-exposed choline-binding (P = 5.00 × 10-08) and helicase proteins (P = 1.32 × 10-06) important for invasion, immune evasion and pneumococcal tropism to CNS. The small effect sizes and negligible heritability indicated that causation of CNS infection requires multiple genetic and other factors reflecting a complex and polygenic aetiology. Our findings suggest that certain pathogen genetic variation modulate pneumococcal survival and tropism to CNS tissue, and therefore, virulence for meningitis.
Collapse
Affiliation(s)
- Chrispin Chaguza
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
- Darwin College, University of Cambridge, Silver Street, Cambridge, UK.
| | - Marie Yang
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Jennifer E Cornick
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Mignon du Plessis
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Rebecca A Gladstone
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Brenda A Kwambana-Adams
- NIHR Global Health Research Unit on Mucosal Pathogens, Division of Infection and Immunity, University College London, London, UK
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Stephanie W Lo
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Chinelo Ebruke
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Gerry Tonkin-Hill
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Chikondi Peno
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- MRC Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Madikay Senghore
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Stephen K Obaro
- Division of Pediatric Infectious Disease, University of Nebraska Medical Center Omaha, Omaha, NE, USA
- International Foundation against Infectious Diseases in Nigeria, Abuja, Nigeria
| | - Sani Ousmane
- Centre de Recherche Médicale et Sanitaire, Niamey, Niger
| | - Gerd Pluschke
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | - Betuel Sigaùque
- Centro de Investigação em Saúde da Manhiça, Maputo, Mozambique
| | - Neil French
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Keith P Klugman
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Robert S Heyderman
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- NIHR Global Health Research Unit on Mucosal Pathogens, Division of Infection and Immunity, University College London, London, UK
| | - Lesley McGee
- Respiratory Diseases Branch, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Martin Antonio
- Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Robert F Breiman
- Emory Global Health Institute, Emory University, Atlanta, GA, USA
| | - Anne von Gottberg
- Centre for Respiratory Diseases and Meningitis, National Institute for Communicable Diseases, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Dean B Everett
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
- MRC Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Aras Kadioglu
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Stephen D Bentley
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
38
|
Feldman C, Anderson R. Platelets and Their Role in the Pathogenesis of Cardiovascular Events in Patients With Community-Acquired Pneumonia. Front Immunol 2020; 11:577303. [PMID: 33042161 PMCID: PMC7527494 DOI: 10.3389/fimmu.2020.577303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Community-acquired pneumonia (CAP) remains an important cause of morbidity and mortality throughout the world with much recent and ongoing research focused on the occurrence of cardiovascular events (CVEs) during the infection, which are associated with adverse short-term and long-term survival. Much of the research directed at unraveling the pathogenesis of these events has been undertaken in the settings of experimental and clinical CAP caused by the dangerous, bacterial respiratory pathogen, Streptococcus pneumoniae (pneumococcus), which remains the most common bacterial cause of CAP. Studies of this type have revealed that although platelets play an important role in host defense against infection, there is also increasing recognition that hyperactivation of these cells contributes to a pro-inflammatory, prothrombotic systemic milieu that contributes to the etiology of CVEs. In the case of the pneumococcus, platelet-driven myocardial damage and dysfunction is exacerbated by the direct cardiotoxic actions of pneumolysin, a major pore-forming toxin of this pathogen, which also acts as potent activator of platelets. This review is focused on the role of platelets in host defense against infection, including pneumococcal infection in particular, and reviews the current literature describing the potential mechanisms by which platelet activation contributes to cardiovascular complications in CAP. This is preceded by an evaluation of the burden of pneumococcal infection in CAP, the clinical features and putative pathogenic mechanisms of the CVE, and concludes with an evaluation of the potential utility of the anti-platelet activity of macrolides and various adjunctive therapies.
Collapse
Affiliation(s)
- Charles Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, Institute of Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
39
|
Interaction of Macrophages and Cholesterol-Dependent Cytolysins: The Impact on Immune Response and Cellular Survival. Toxins (Basel) 2020; 12:toxins12090531. [PMID: 32825096 PMCID: PMC7551085 DOI: 10.3390/toxins12090531] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 02/07/2023] Open
Abstract
Cholesterol-dependent cytolysins (CDCs) are key virulence factors involved in many lethal bacterial infections, including pneumonia, necrotizing soft tissue infections, bacterial meningitis, and miscarriage. Host responses to these diseases involve myeloid cells, especially macrophages. Macrophages use several systems to detect and respond to cholesterol-dependent cytolysins, including membrane repair, mitogen-activated protein (MAP) kinase signaling, phagocytosis, cytokine production, and activation of the adaptive immune system. However, CDCs also promote immune evasion by silencing and/or destroying myeloid cells. While there are many common themes between the various CDCs, each CDC also possesses specific features to optimally benefit the pathogen producing it. This review highlights host responses to CDC pathogenesis with a focus on macrophages. Due to their robust plasticity, macrophages play key roles in the outcome of bacterial infections. Understanding the unique features and differences within the common theme of CDCs bolsters new tools for research and therapy.
Collapse
|
40
|
Nishimoto AT, Rosch JW, Tuomanen EI. Pneumolysin: Pathogenesis and Therapeutic Target. Front Microbiol 2020; 11:1543. [PMID: 32714314 PMCID: PMC7343714 DOI: 10.3389/fmicb.2020.01543] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/15/2020] [Indexed: 01/08/2023] Open
Abstract
Streptococcus pneumoniae is an opportunistic pathogen responsible for widespread illness and is a major global health issue for children, the elderly, and the immunocompromised population. Pneumolysin (PLY) is a cholesterol-dependent cytolysin (CDC) and key pneumococcal virulence factor involved in all phases of pneumococcal disease, including transmission, colonization, and infection. In this review we cover the biology and cytolytic function of PLY, its contribution to S. pneumoniae pathogenesis, and its known interactions and effects on the host with regard to tissue damage and immune response. Additionally, we review statins as a therapeutic option for CDC toxicity and PLY toxoid as a vaccine candidate in protein-based vaccines.
Collapse
Affiliation(s)
- Andrew T Nishimoto
- Department of Infectious Disease, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Jason W Rosch
- Department of Infectious Disease, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Elaine I Tuomanen
- Department of Infectious Disease, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
41
|
Thanawastien A, Joyce KE, Cartee RT, Haines LA, Pelton SI, Tweten RK, Killeen KP. Preclinical in vitro and in vivo profile of a highly-attenuated, broadly efficacious pneumolysin genetic toxoid. Vaccine 2020; 39:1652-1660. [PMID: 32532546 PMCID: PMC8237519 DOI: 10.1016/j.vaccine.2020.04.064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 04/03/2020] [Accepted: 04/26/2020] [Indexed: 02/05/2023]
Abstract
Pneumolysin is a highly conserved, cholesterol-dependent cytolysin that is an important Streptococcus pneumoniae virulence factor and an attractive target for vaccine development. To attenuate pneumolysin toxicity, a genetic toxoid was constructed with two amino acid changes, G293S and L460D, termed PLY-D, that reduced cytolytic activity > 125,000-fold. In mice, PLY-D elicited high anti-PLY IgG antibody titers that neutralized the cytolytic activity of the wild-type toxin in vitro. To evaluate the protective efficacy of PLY-D, mice were immunized intramuscularly and then challenged intranasally with a lethal dose of 28 clinical isolates of S. pneumoniae originating from different geographical locations, disease states (i.e. bacteremia, pneumonia), or body sites (i.e. sputum, blood). PLY-D immunization conferred significant protection from challenge with 17 of 20 serotypes (85%) and 22 of 28 strains (79%). Further, we demonstrated that immunization with PLY-D provided statistically significant improvement in survival against challenge with serotype 4 and 18C strains compared to mice immunized with a pneumococcal conjugate vaccine Prevnar 13® (PCV13). Co-administration of PLY-D and PCV13 conferred greater protection against challenge with a serotype 6B strain than immunization with either vaccine alone. These data indicate that PLY-D is a broadly protective antigen with the potential to serve as a serotype-independent vaccine against invasive pneumococcal disease either alone or in combination with PCVs.
Collapse
Affiliation(s)
- Ann Thanawastien
- Matrivax Research & Development Corporation, Boston, MA, United States
| | - Kelsey E Joyce
- Matrivax Research & Development Corporation, Boston, MA, United States
| | - Robert T Cartee
- Matrivax Research & Development Corporation, Boston, MA, United States
| | - Laurel A Haines
- Matrivax Research & Development Corporation, Boston, MA, United States
| | - Stephen I Pelton
- Boston University Schools of Medicine and Public Health, Boston Medical Center, Boston, MA, United States
| | - Rodney K Tweten
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Kevin P Killeen
- Matrivax Research & Development Corporation, Boston, MA, United States.
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW The human body plays host to bacterial biofilms across diverse anatomical sites. The treatment of pathogenic biofilm infection is confounded by their high rate of antibiotic resistance. Therefore, it is critical to understand the interplay between these biofilms and the host immune system to develop new tactics to combat these infections. RECENT FINDINGS Bacterial biofilms and the components they produce affect and are affected by the host immune system. Host anatomical sites represent distinct niches in which defined bacterial biofilms are able to form and interact with the host immune system. For persistent colonization to occur, the bacteria must either avoid or suppress the host immune system, or induce an immune response that facilitates their perpetuation. SUMMARY Commensal bacterial biofilms form a protective barrier against colonization by pathogens. Using similar mechanisms, bacteria modulate the immune system to orchestrate persistence and sometimes disease. Clinicians must balance the need to avoid disturbing beneficial commensal biofilms with the difficulty in preventing or treating pathogenic bacterial biofilms such as those that develop on medical implants and open wounds.
Collapse
Affiliation(s)
- Christina N Morra
- Department of Microbiology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
43
|
Feldman C. Cardiac complications in community-acquired pneumonia and COVID-19. Afr J Thorac Crit Care Med 2020; 26:10.7196/AJTCCM.2020.v26i2.077. [PMID: 34235421 PMCID: PMC7221539 DOI: 10.7196/ajtccm.2020.v26i2.077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
Community-acquired pneumonia (CAP) remains a global health problem with significant morbidity and mortality. Much recent published literature about the infection has indicated that a substantial number of patients with CAP, particularly those ill enough to be admitted to hospital, will suffer a cardiovascular event. While these may include events such as deep venous thrombosis and stroke, most of the events involve the heart and include the occurrence of an arrhythmia (most commonly atrial fibrillation), new onset or worsening of heart failure and acute myocardial infarction. While such cardiac events may occur, for example, in all-cause CAP and CAP due to influenza virus infection, and more recently described with the SARS-CoV-2 pandemic, a significant amount of research work has been investigating the pathogenic mechanisms of these cardiac events in patients with CAP due to Streptococcus pneumoniae (pneumococcus) and, more recently, COVID-19 infections. Such research has identified a number of mechanisms by which these microorganisms may cause cardiovascular events. Importantly, these cardiac events appear not only to be associated with in-hospital mortality, but they also appear to contribute to longer-term mortality of patients with CAP, even after their discharge from hospital. This review will focus initially on studies of cardiovascular events in all-cause CAP and pneumococcal CAP, excluding COVID-19 infection, and then address similar issues in the latter infection.
Collapse
Affiliation(s)
- C Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
44
|
Bo Y, Jiang Y, Chen K, Cai K, Li W, Roy J, Bao Y, Cheng J. Targeting infected host cells in vivo via responsive azido-sugar mediated metabolic cell labeling followed by click reaction. Biomaterials 2020; 238:119843. [DOI: 10.1016/j.biomaterials.2020.119843] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/02/2020] [Accepted: 02/03/2020] [Indexed: 01/24/2023]
|
45
|
Streptococcus pneumoniae Elaborates Persistent and Prolonged Competent State during Pneumonia-Derived Sepsis. Infect Immun 2020; 88:IAI.00919-19. [PMID: 31988172 DOI: 10.1128/iai.00919-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 01/21/2020] [Indexed: 01/18/2023] Open
Abstract
The competence regulon of pneumococcus regulates both genetic transformation and virulence. However, competence induction during host infection has not been examined. By using the serotype 2 strain D39, we transcriptionally fused the firefly luciferase (luc) to competence-specific genes and spatiotemporally monitored the competence development in a mouse model of pneumonia-derived sepsis. In contrast to the universally reported short transient burst of competent state in vitro, the naturally developed competent state was prolonged and persistent during pneumonia-derived sepsis. The competent state began at approximately 20 h postinfection (hpi) and facilitated systemic invasion and sepsis development and progressed in different manners. In some mice, acute pneumonia quickly led to sepsis and death, accompanied by increasing intensity of the competence signal. In the remaining mice, pneumonia lasted longer, with the competence signal decreasing at first but increasing as the infection became septic. The concentration of pneumococcal inoculum (1 × 106 to 1 × 108 CFU/mouse) and postinfection lung bacterial burden did not appreciably impact the kinetics of competence induction. Exogenously provided competence stimulating peptide 1 (CSP1) failed to modulate the onset kinetics of competence development in vivo The competence shutoff regulator DprA was highly expressed during pneumonia-derived sepsis but failed to turn off the competent state in mice. Competent D39 bacteria propagated the competence signal through cell-to-cell contact rather than the classically described quorum-sensing mechanism. Finally, clinical pneumococcal strains of different serotypes were also able to develop natural competence during pneumonia-derived sepsis.
Collapse
|
46
|
Chiong YK, Tan I, Lim TC, Thong WY, Chan MY, Chan SM. Pneumococcal Pneumonia Resembling Acute Myocardial Infarction in an Adolescent Male. Pediatr Infect Dis J 2020; 39:81-84. [PMID: 31725548 DOI: 10.1097/inf.0000000000002527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Myocarditis is commonly associated with viruses. However, we present a novel case of a teenager with pneumococcal pneumonia and bacteremia complicated by myocarditis and rhabdomyolysis, presenting with features of acute ST-elevation myocardial infarction and cardiogenic shock. Physicians should be aware that Streptococcus pneumoniae infection, like Group A Streptococcus, can mimic acute myocardial infarction in young males without cardiovascular risk factors.
Collapse
Affiliation(s)
- Yee Keow Chiong
- From the Khoo Teck Puat National University Children's Medical Institute, National University Health System, Singapore
| | - Ian Tan
- From the Khoo Teck Puat National University Children's Medical Institute, National University Health System, Singapore
| | - Terence Cw Lim
- From the Khoo Teck Puat National University Children's Medical Institute, National University Health System, Singapore
| | - Wen Yi Thong
- From the Khoo Teck Puat National University Children's Medical Institute, National University Health System, Singapore
| | - Mark Y Chan
- Department of Cardiology, National University Hospital, Singapore
| | - Si Min Chan
- From the Khoo Teck Puat National University Children's Medical Institute, National University Health System, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
47
|
Adams W, Bhowmick R, Bou Ghanem EN, Wade K, Shchepetov M, Weiser JN, McCormick BA, Tweten RK, Leong JM. Pneumolysin Induces 12-Lipoxygenase-Dependent Neutrophil Migration during Streptococcus pneumoniae Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:101-111. [PMID: 31776202 PMCID: PMC7195902 DOI: 10.4049/jimmunol.1800748] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/16/2019] [Indexed: 12/23/2022]
Abstract
Streptococcus pneumoniae is a major cause of pneumonia, wherein infection of respiratory mucosa drives a robust influx of neutrophils. We have previously shown that S. pneumoniae infection of the respiratory epithelium induces the production of the 12-lipoxygenase (12-LOX)-dependent lipid inflammatory mediator hepoxilin A3, which promotes recruitment of neutrophils into the airways, tissue damage, and lethal septicemia. Pneumolysin (PLY), a member of the cholesterol-dependent cytolysin (CDC) family, is a major S. pneumoniae virulence factor that generates ∼25-nm diameter pores in eukaryotic membranes and promotes acute inflammation, tissue damage, and bacteremia. We show that a PLY-deficient S. pneumoniae mutant was impaired in triggering human neutrophil transepithelial migration in vitro. Ectopic production of PLY endowed the nonpathogenic Bacillus subtilis with the ability to trigger neutrophil recruitment across human-cultured monolayers. Purified PLY, several other CDC family members, and the α-toxin of Clostridium septicum, which generates pores with cross-sectional areas nearly 300 times smaller than CDCs, reproduced this robust neutrophil transmigration. PLY non-pore-forming point mutants that are trapped at various stages of pore assembly did not recruit neutrophils. PLY triggered neutrophil recruitment in a 12-LOX-dependent manner in vitro. Instillation of wild-type PLY but not inactive derivatives into the lungs of mice induced robust 12-LOX-dependent neutrophil migration into the airways, although residual inflammation induced by PLY in 12-LOX-deficient mice indicates that 12-LOX-independent pathways also contribute to PLY-triggered pulmonary inflammation. These data indicate that PLY is an important factor in promoting hepoxilin A3-dependent neutrophil recruitment across pulmonary epithelium in a pore-dependent fashion.
Collapse
Affiliation(s)
- Walter Adams
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192
| | - Rudra Bhowmick
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111
| | - Elsa N Bou Ghanem
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111
| | - Kristin Wade
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Mikhail Shchepetov
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104
| | - Jeffrey N Weiser
- Department of Microbiology, New York University School of Medicine, New York, NY 10016; and
| | - Beth A McCormick
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655
| | - Rodney K Tweten
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - John M Leong
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111;
| |
Collapse
|
48
|
Lv Q, Zhang P, Quan P, Cui M, Liu T, Yin Y, Chi G. Quercetin, a pneumolysin inhibitor, protects mice against Streptococcus pneumoniae infection. Microb Pathog 2019; 140:103934. [PMID: 31862394 DOI: 10.1016/j.micpath.2019.103934] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/14/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022]
Abstract
Pneumolysin (PLY), a pore-forming cytotoxin and a major virulence determinant, is a member of the cholesterol-dependent cytolysin (CDC) family and essential for promoting Streptococcus pneumoniae (S.pneumoniae) infection. Due to the action characteristics of hemolysin itself, the pneumolysin released after killing bacteria with conventional antibiotics still has the ability to damage host cells; therefore, drug treatments directly inhibiting hemolysin activity are the most effective. Hemolysis assays were used to confirm that quercetin can inhibit the activity of PLY, protecting cells in vitro, and an oligomerization assay was used to determine the mechanism of quercetin to suppress PLY activity. Live/Dead testing, lactate dehydrogenase (LDH) release analysis and a murine model of endonasal pulmonary infection were used to explore the capability of quercetin to protect cells and mice from S. pneumoniae-mediated damage in vivo and in vitro. The results indicated that quercetin significantly reduced PLY-induced hemolytic activity and cytotoxicity via repressing the formation of oligomers. In addition, treatment with quercetin can reduce PLY-mediated cell injury, improve the survival rate of mice infected with a lethal dose of S. pneumoniae, alleviate the pathological damage of lung tissue and inhibit the release of cytokines (IL-1β and TNF-α) in bronchoalveolar lavage fluid. Considering the importance of these events in antimicrobial resistant S. pneumoniae pathogenesis, our results indicated that quercetin may be a novel potential drug candidate for the treatment of clinical pneumococcal infections.
Collapse
Affiliation(s)
- Qianghua Lv
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China; Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Peng Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Pusheng Quan
- Department of Neurology, The First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Mengyao Cui
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Tianjiao Liu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Yunhou Yin
- School of Communication, Guizhou Minzu University, Guiyang, China
| | - Gefu Chi
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
49
|
Opening the OPK Assay Gatekeeper: Harnessing Multi-Modal Protection by Pneumococcal Vaccines. Pathogens 2019; 8:pathogens8040203. [PMID: 31652741 PMCID: PMC6963391 DOI: 10.3390/pathogens8040203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 10/14/2019] [Accepted: 10/21/2019] [Indexed: 11/16/2022] Open
Abstract
Pneumococcal vaccine development is driven by the achievement of high activity in a single gatekeeper assay: the bacterial opsonophagocytic killing (OPK) assay. New evidence challenges the dogma that anti-capsular antibodies have only a single function that predicts success. The emerging concept of multi-modal protection presents an array of questions that are fundamental to adopting a new vaccine design process. If antibodies have hidden non-opsonic functions that are protective, should these be optimized for better vaccines? What would protein antigens add to protective activity? Are cellular immune functions additive to antibodies for success? Do different organs benefit from different modes of protection? Can vaccine activities beyond OPK protect the immunocompromised host? This commentary raises these issues at a time when capsule-only OPK assay-based vaccines are increasingly seen as a limiting strategy.
Collapse
|
50
|
|