Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
2.
Cureus ; 15(4): e37696, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37206519

ABSTRACT

Nitrous oxide (N2O) misuse creates a diagnostic dilemma due to its clinical presentation, difficulty in identification, and toxicity related to its chronic abuse, with resultant morbidity and mortality. Chronic abuse can lead to myeloneuropathy and subacute combined degeneration in otherwise healthy individuals. Health professionals should be aware of the commercial availability and abuse of N2O by the public, and N2O toxicity should be included in the differential diagnosis in patients presenting with myelopathy of unknown etiology. A case report was conducted on a 38-year-old female at approximately 30 weeks of gestation who presented to the emergency department with worsening bilateral lower extremity numbness, tingling, and weakness. The patient admitted to nitrous oxide inhalation during the two months prior to admission. She reported using four cans of whippets per week (approximately 8 g of N2O per whippet) up to 50 cans per day (400 g N2O) prior to the onset of symptoms. An MRI of the cervical spine was performed, showing T2 hyperintensity from C2 to C6 involving dorsal columns indicative of subacute combined degeneration. The patient was treated with intravenous vitamin B12 due to the clinical and radiographic evidence of nitrous oxide-induced myelopathy. The pathophysiology of N2O toxicity involves the oxidation of the cobalt atom of cobalamin (vitamin B12) from its reduced active 1+ valent state to its oxidized inactive 3+ valent state. This oxidation inactivates the enzyme methionine synthetase. B12 is an essential cofactor for downstream DNA synthesis. Consequently, excess N2O creates functional B12 deficiency leading to irreversible nerve damage if left undiagnosed and untreated.

3.
J Psychosoc Nurs Ment Health Serv ; 61(8): 17-24, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36853038

ABSTRACT

Health care personnel who have close, face-to-face patient contact experience more workplace violence (WPV) than employees in other fields. Certain health care departments (i.e., high-incidence care areas) have elevated rates of WPV that can have adverse emotional, physical, and financial consequences for patients, employees, and institutions. Health care workers need de-escalation training to efficiently manage patient aggression while also safeguarding patients' dignity and patient-provider trust. The current Plan, Do, Study, Act quality improvement project used insights from an in-depth literature review to create a 1-hour, evidence-based, in-service de-escalation training for personnel from high-incidence care areas. A pre/post design was used to evaluate participants' responses to the Confidence Coping with Patient Aggression Instrument. Post-training, participants reported significantly increased feelings of safety regarding potential patient aggression (p = 0.001) and more efficacy regarding their aggression management techniques (p = 0.039). Based on the training's results, recommendations were made for future institutional de-escalation initiatives. [Journal of Psychosocial Nursing and Mental Health Services, 61(8), 17-24.].


Subject(s)
Aggression , Workplace Violence , Humans , Incidence , Aggression/psychology , Workplace Violence/prevention & control , Workplace Violence/psychology , Patients , Health Personnel/education
4.
mSphere ; 7(3): e0013022, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35642538

ABSTRACT

Nonhealing diabetic foot ulcers (DFU), a major complication of diabetes, are associated with high morbidity and mortality despite current standard of care. Since Staphylococcus aureus is the most common pathogen isolated from nonhealing and infected DFU, we hypothesized that S. aureus virulence factors would damage tissue, promote immune evasion and alter the microbiome, leading to bacterial persistence and delayed wound healing. In a diabetic mouse polymicrobial wound model with S. aureus, Pseudomonas aeruginosa, and Streptococcus pyogenes, we report a rapid bacterial proliferation, prolonged pro-inflammatory response and large necrotic lesions unclosed for up to 40 days. Treatment with AZD6389, a three-monoclonal antibody combination targeting S. aureus alpha toxin, 4 secreted leukotoxins, and fibrinogen binding cell-surface adhesin clumping factor A resulted in full skin re-epithelization 21 days after inoculation. By neutralizing multiple virulence factors, AZD6389 effectively blocked bacterial agglutination and S. aureus-mediated cell killing, abrogated S. aureus-mediated immune evasion and targeted the bacteria for opsonophagocytic killing. Neutralizing S. aureus virulence not only facilitated S. aureus clearance in lesions, but also reduced S. pyogenes and P. aeruginosa numbers, damaging inflammatory mediators and markers for neutrophil extracellular trap formation 14 days post initiation. Collectively, our data suggest that AZD6389 holds promise as an immunotherapeutic approach against DFU complications. IMPORTANCE Diabetic foot ulcers (DFU) represent a major complication of diabetes and are associated with poor quality of life and increased morbidity and mortality despite standard of care. They have a complex pathogenesis starting with superficial skin lesions, which often progress to deeper tissue structures up to the bone and ultimately require limb amputation. The skin microbiome of diabetic patients has emerged as having an impact on DFU occurrence and chronicity. DFU are mostly polymicrobial, and the Gram-positive bacterium Staphylococcus aureus detected in more than 95% of cases. S. aureus possess a collection of virulence factors which participate in disease progression and may facilitate growth of other pathogens. Here we show in a diabetic mouse wound model that targeting some specific S. aureus virulence factors with a multimechanistic antibody combination accelerated wound closure and promoted full skin re-epithelization. This work opens promising new avenues for the treatment of DFU.


Subject(s)
Diabetes Mellitus , Diabetic Foot , Staphylococcal Infections , Animals , Antibodies, Monoclonal , Bacteria , Diabetic Foot/complications , Diabetic Foot/drug therapy , Mice , Pseudomonas aeruginosa , Quality of Life , Staphylococcal Infections/microbiology , Staphylococcus aureus , Virulence , Virulence Factors
5.
Cells ; 10(4)2021 04 09.
Article in English | MEDLINE | ID: mdl-33918759

ABSTRACT

Impaired DNA damage responses are associated with several diseases, including pregnancy complications. Recent research identified an ATM-kinase dependent function for the nuclear isoform of the receptor for advanced glycation end-products (RAGE) during double strand break (DSB)-repair. RAGE contributes to end-resectioning of broken DNA sites by binding with the MRE11-Rad50-Nbs1 (MRN) complex. Placental research is limited regarding the impact of genomic instability and the mechanism for potential repair. We tested the hypothesis regarding the involvement of RAGE during the repair of placental DNA-DSBs. We first identified that the pregnancy complications of PE and preterm labor (PTL) experience loss of genomic integrity and an in vitro trophoblast cell model was used to characterize trophoblast DSBs. Colocalized immunofluorescence of γ-H2AX and RAGE support the potential involvement of RAGE in cellular responses to DNA-DSBs. Immunoblotting for both molecules in PE and PTL placenta samples and in trophoblast cells validated a connection. Co-immunoprecipitation studies revealed interactions between RAGE and pATM and MRE11 during DNA-DSBs. Reduced cellular invasion confirmed the role of genomic instability in trophoblastic function. Collectively, these experiments identified genomic instability in pregnancy complications, the impact of defective DNA on trophoblast function, and a possible RAGE-mediated mechanism during DNA-DSB repair.


Subject(s)
DNA Breaks, Double-Stranded , Receptor for Advanced Glycation End Products/metabolism , Trophoblasts/metabolism , Trophoblasts/pathology , Adult , Ataxia Telangiectasia Mutated Proteins/metabolism , Bleomycin , Case-Control Studies , Cell Line , Cell Nucleus/metabolism , Female , Genomic Instability , Histones/metabolism , Humans , MRE11 Homologue Protein/metabolism , Pregnancy , Pregnancy Complications/genetics , Protein Binding , Tobacco Products
6.
Article in English | MEDLINE | ID: mdl-32152087

ABSTRACT

Antibiotics revolutionized the treatment of infectious diseases; however, it is now clear that broad-spectrum antibiotics alter the composition and function of the host's microbiome. The microbiome plays a key role in human health, and its perturbation is increasingly recognized as contributing to many human diseases. Widespread broad-spectrum antibiotic use has also resulted in the emergence of multidrug-resistant pathogens, spurring the development of pathogen-specific strategies such as monoclonal antibodies (MAbs) to combat bacterial infection. Not only are pathogen-specific approaches not expected to induce resistance in nontargeted bacteria, but they are hypothesized to have minimal impact on the gut microbiome. Here, we compare the effects of antibiotics, pathogen-specific MAbs, and their controls (saline or control IgG [c-IgG]) on the gut microbiome of 7-week-old, female, C57BL/6 mice. The magnitude of change in taxonomic abundance, bacterial diversity, and bacterial metabolites, including short-chain fatty acids (SCFA) and bile acids in the fecal pellets from mice treated with pathogen-specific MAbs, was no different from that with animals treated with saline or an IgG control. Conversely, dramatic changes were observed in the relative abundance, as well as alpha and beta diversity, of the fecal microbiome and bacterial metabolites in the feces of all antibiotic-treated mice. Taken together, these results indicate that pathogen-specific MAbs do not alter the fecal microbiome like broad-spectrum antibiotics and may represent a safer, more-targeted approach to antibacterial therapy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Gastrointestinal Microbiome/drug effects , Animals , Bile Acids and Salts/metabolism , DNA, Bacterial/analysis , Fatty Acids/metabolism , Feces/microbiology , Female , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics , Specific Pathogen-Free Organisms
7.
J Clin Invest ; 129(5): 2133-2144, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30985291

ABSTRACT

Diabetic individuals are at considerable risk for invasive infection by Staphylococcus aureus, however, the mechanisms underlying this enhanced susceptibility to infection are unclear. We observed increased mortality following i.v. S. aureus infection in diabetic mice compared with nondiabetic controls, correlating with increased numbers of low-density neutrophils (LDNs) and neutrophil extracellular traps (NETs). LDNs have been implicated in the inflammatory pathology of diseases such as lupus, given their release of large amounts of NETs. Our goal was to describe what drives LDN increases during S. aureus infection in the diabetic host and mechanisms that promote increased NET production by LDNs. LDN development is dependent on TGF-ß, which we found to be more activated in the diabetic host. Neutralization of TGF-ß, or the TGF-ß-activating integrin αvß8, reduced LDN numbers and improved survival during S. aureus infection. Targeting S. aureus directly with MEDI4893*, an α toxin-neutralizing monoclonal antibody, blocked TGF-ß activation, reduced LDNs and NETs, and significantly improved survival. A comparison of gene and protein expression in high-density neutrophils and LDNs identified increased GPCRs and elevated phosphatase and tensin homolog (PTEN) in the LDN subset. Inhibition of PTEN improved the survival of infected diabetic mice. Our data identify a population of neutrophils in infected diabetic mice that correlated with decreased survival and increased NET production and describe 3 therapeutic targets, a bacterial target and 2 host proteins, that prevented NET production and improved survival.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Broadly Neutralizing Antibodies/pharmacology , Extracellular Traps/immunology , Neutrophils/cytology , Neutrophils/microbiology , Staphylococcal Infections/immunology , Staphylococcus aureus , Animals , Cell Separation , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/immunology , Disease Models, Animal , Female , Flow Cytometry , Immunoglobulin G/metabolism , Inflammation , Integrins/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Risk Factors , Signal Transduction , Staphylococcal Infections/complications , Streptozocin , Transforming Growth Factor beta/metabolism
8.
Am J Respir Cell Mol Biol ; 59(6): 745-756, 2018 12.
Article in English | MEDLINE | ID: mdl-30109945

ABSTRACT

Pseudomonas aeruginosa and Klebsiella pneumoniae are two common gram-negative pathogens that are associated with bacterial pneumonia and can often be isolated from the same patient. We used a mixed-pathogen pneumonia infection model in which mice were infected with sublethal concentrations of P. aeruginosa and K. pneumoniae, resulting in significant lethality, outgrowth of both bacteria in the lung, and systemic dissemination of K. pneumoniae. Inflammation, induced by P. aeruginosa activation of Toll-like receptor 5, results in prolonged neutrophil recruitment to the lung and increased levels of neutrophil elastase in the airway, resulting in lung damage and epithelial barrier dysfunction. Live P. aeruginosa was not required to potentiate K. pneumoniae infection, and flagellin alone was sufficient to induce lethality when delivered along with Klebsiella. Prophylaxis with an anti-Toll-like receptor 5 antibody or Sivelestat, a neutrophil elastase inhibitor, reduced neutrophil influx, inflammation, and mortality. Furthermore, pathogen-specific monoclonal antibodies targeting P. aeruginosa or K. pneumoniae prevented the outgrowth of both bacteria and reduced host inflammation and lethality. These findings suggest that coinfection with P. aeruginosa may enable the outgrowth and dissemination of K. pneumoniae, and that a pathogen- or host-specific prophylactic approach targeting P. aeruginosa may prevent or limit the severity of such infections by reducing neutrophil-induced lung damage.


Subject(s)
Coinfection/immunology , Klebsiella Infections/immunology , Klebsiella pneumoniae/immunology , Neutrophils/immunology , Pneumonia/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Animals , Cells, Cultured , Coinfection/microbiology , Coinfection/pathology , Female , Klebsiella Infections/microbiology , Klebsiella Infections/pathology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Neutrophils/microbiology , Neutrophils/pathology , Pneumonia/microbiology , Pneumonia/pathology , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Toll-Like Receptor 5/metabolism
9.
Cell Rep ; 22(9): 2431-2441, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29490278

ABSTRACT

Clinical severity of Staphylococcus aureus respiratory infection correlates with alpha toxin (AT) expression. AT activates the NLRP3 inflammasome; deletion of Nlrp3, or AT neutralization, protects mice from lethal S. aureus pneumonia. We tested the hypothesis that this protection is not due to a reduction in inflammasome-dependent cytokines (IL-1ß/IL-18) but increased bactericidal function of macrophages. In vivo, neutralization of AT or NLRP3 improved bacterial clearance and survival, while blocking IL-1ß/IL-18 did not. Primary human monocytes were used in vitro to determine the mechanism through which NLRP3 alters bacterial killing. In cells treated with small interfering RNA (siRNA) targeting NLRP3 or infected with AT-null S. aureus, mitochondria co-localize with bacterial-containing phagosomes. Mitochondrial engagement activates caspase-1, a process dependent on complex II of the electron transport chain, near the phagosome, promoting its acidification. These data demonstrate a mechanism utilized by S. aureus to sequester itself from antimicrobial processes within the cell.


Subject(s)
Immune Evasion , Macrophages/microbiology , Microbial Viability , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Staphylococcus aureus/metabolism , Animals , Bacterial Toxins , Caspase 1/metabolism , Electron Transport Complex II/metabolism , Female , Hemolysin Proteins , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Monocytes/metabolism , Neutralization Tests , Protein Transport , Reactive Oxygen Species/metabolism
10.
Article in English | MEDLINE | ID: mdl-28584141

ABSTRACT

Secreted alpha-toxin and surface-localized clumping factor A (ClfA) are key virulence determinants in Staphylococcus aureus bloodstream infections. We previously demonstrated that prophylaxis with a multimechanistic monoclonal antibody (MAb) combination against alpha-toxin (MEDI4893*) and ClfA (11H10) provided greater strain coverage and improved efficacy in an S. aureus lethal bacteremia model. Subsequently, 11H10 was found to exhibit reduced affinity and impaired inhibition of fibrinogen binding to ClfA002 expressed by members of a predominant hospital-associated methicillin-resistant S. aureus (MRSA) clone, ST5. Consequently, we identified another anti-ClfA MAb (SAR114) from human tonsillar B cells with >100-fold increased affinity for three prominent ClfA variants, including ClfA002, and potent inhibition of bacterial agglutination by 112 diverse clinical isolates. We next constructed bispecific Abs (BiSAbs) comprised of 11H10 or SAR114 as IgG scaffolds and grafted anti-alpha-toxin (MEDI4893*) single-chain variable fragment to the amino or carboxy terminus of the anti-ClfA heavy chains. Although the BiSAbs exhibited in vitro potencies similar to those of the parental MAbs, only 11H10-BiSAb, but not SAR114-BiSAb, showed protective activity in murine infection models comparable to the respective MAb combination. In vivo activity with SAR114-BiSAb was observed in infection models with S. aureus lacking ClfA. Our data suggest that high-affinity binding to ClfA sequesters the SAR114-BiSAb to the bacterial surface, thereby reducing both alpha-toxin neutralization and protection in vivo These results indicate that a MAb combination targeting ClfA and alpha-toxin is more promising for future development than the corresponding BiSAb.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Bacteremia/drug therapy , Bacterial Toxins/immunology , Coagulase/immunology , Hemolysin Proteins/immunology , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Animals , Antibodies, Bacterial/immunology , Antibodies, Bacterial/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/immunology , Bacteremia/microbiology , Broadly Neutralizing Antibodies , Female , Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Staphylococcal Infections/immunology , Virulence Factors
11.
Sci Rep ; 6: 35466, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27739519

ABSTRACT

Bacterial pneumonia, such as those caused by Staphylococcus aureus, is associated with an influx of inflammatory neutrophils into the lung tissue and airways. Regulation and clearance of recruited neutrophils is essential for preventing tissue damage by "friendly fire", a responsibility of macrophages in a process called efferocytosis. We hypothesized that S. aureus impairs efferocytosis by alveolar macrophages (AMs) through the activity of the secreted virulence factor alpha toxin (AT), which has been implicated in altering the antimicrobial function of AMs. Infection of mice lacking AMs resulted in significantly increased numbers of neutrophils in the lung, while clearance of neutrophils delivered intranasally into uninfected mice was reduced in AM depleted animals. In vitro, sublytic levels of AT impaired uptake of apoptotic neutrophils by purified AMs. In vivo, the presence of AT reduced uptake of neutrophils by AMs. Differential uptake of neutrophils was not due to changes in either the CD47/CD172 axis or CD36 levels. AT significantly reduced lung expression of CCN1 and altered AM surface localization of DD1α, two proteins known to influence efferocytosis. We conclude that AT may contribute to tissue damage during S. aureus pneumonia by inhibiting the ability of AM to clear neutrophils at the site of infection.


Subject(s)
Macrophages/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Neutrophils/immunology , Pneumonia, Bacterial/immunology , Virulence Factors/toxicity , Animals , CD36 Antigens/genetics , CD36 Antigens/metabolism , CD47 Antigen/genetics , CD47 Antigen/metabolism , Cell Movement , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Female , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neutrophils/physiology , Pneumonia, Bacterial/microbiology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism
12.
Sci Transl Med ; 8(329): 329ra31, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26962155

ABSTRACT

Broad-spectrum antibiotic use may adversely affect a patient's beneficial microbiome and fuel cross-species spread of drug resistance. Although alternative pathogen-specific approaches are rationally justified, a major concern for this precision medicine strategy is that co-colonizing or co-infecting opportunistic bacteria may still cause serious disease. In a mixed-pathogen lung infection model, we find that the Staphylococcus aureus virulence factor α toxin potentiates Gram-negative bacterial proliferation, systemic spread, and lethality by preventing acidification of bacteria-containing macrophage phagosomes, thereby reducing effective killing of both S. aureus and Gram-negative bacteria. Prophylaxis or early treatment with a single α toxin neutralizing monoclonal antibody prevented proliferation of co-infecting Gram-negative pathogens and lethality while also promoting S. aureus clearance. These studies suggest that some pathogen-specific, antibody-based approaches may also work to reduce infection risk in patients colonized or co-infected with S. aureus and disparate drug-resistant Gram-negative bacterial opportunists.


Subject(s)
Bacterial Toxins/adverse effects , Hemolysin Proteins/adverse effects , Opportunistic Infections/microbiology , Respiratory Tract Infections/microbiology , Staphylococcal Infections/microbiology , Acids/metabolism , Animals , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Calpain/metabolism , Coinfection/microbiology , Enzyme Activation/drug effects , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Lysosomes/metabolism , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/pathology , Mice , Microbial Viability/drug effects , Models, Biological , Neutrophils/drug effects , Neutrophils/pathology , Opportunistic Infections/pathology , Pneumonia/microbiology , Pneumonia/pathology , Pseudomonas aeruginosa/drug effects , Respiratory Tract Infections/pathology , Staphylococcal Infections/pathology , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development
13.
Antimicrob Agents Chemother ; 59(1): 299-309, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25348518

ABSTRACT

Alpha-toxin (AT) is a major virulence determinant in Staphylococcus aureus skin and soft tissue infection models. We previously demonstrated that prophylactic administration of 2A3, an AT-neutralizing monoclonal antibody (MAb), prevents S. aureus disease in a mouse dermonecrosis model by neutralizing AT-mediated tissue necrosis and immune evasion. In the present study, MEDI4893*, an affinity-optimized version of 2A3, was characterized for therapeutic activity in the dermonecrosis model as a single agent and in combination with two frontline antibiotics, vancomycin and linezolid. MEDI4893* postinfection therapy was found to exhibit a therapeutic treatment window similar to that for linezolid but longer than that for vancomycin. Additionally, when combined with either vancomycin or linezolid, MEDI4893* resulted in reduced tissue damage, increased neutrophil and macrophage infiltration and abscess formation, and accelerated healing relative to those with the antibiotic monotherapies. These data suggest that AT neutralization with a potent MAb holds promise for both prophylaxis and adjunctive therapy with antibiotics and may be a valuable addition to currently available options for the treatment of S. aureus skin and soft tissue infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Bacterial Toxins/immunology , Hemolysin Proteins/immunology , Staphylococcal Skin Infections/drug therapy , Animals , Anti-Bacterial Agents/pharmacokinetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Broadly Neutralizing Antibodies , Disease Models, Animal , Drug Therapy, Combination , Female , Linezolid/pharmacokinetics , Linezolid/pharmacology , Mice, Inbred BALB C , Necrosis/drug therapy , Necrosis/microbiology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Vancomycin/pharmacokinetics , Vancomycin/pharmacology
14.
Parasitol Res ; 109(3): 675-87, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21369763

ABSTRACT

The intimate host-parasite relationship mandates adaptation to the genetic and phenotypic variability of their counterparts. Here, inbred and outcrossed strains of Schistosoma mansoni were challenged with "local" and "novel" intermediate and definitive hosts to examine effects of genetic variability and novelty on infection success and dynamics. Genetically distinct lines of Biomphalaria glabrata intermediate hosts exposed to inbred and outcrossed S. mansoni larvae were assessed for differences in both snail and parasite life-history parameters. Cercariae from each parasite-snail treatment were used to infect "local" and "novel" Mus musculus definitive hosts to assess parasite infectivity and fitness. Outcrossed parasites significantly reduced snail growth, were more productive, and induced greater host mortality than inbred parasites. Mouse strain did not influence parasite infectivity or reproduction, but parasite and snail host genetic background did, affecting both sex-specific infectivity and parasite productivity. Overall, genetic background of S. mansoni and its intermediate snail host altered life history traits and transmission dynamics of the parasite throughout its life cycle.


Subject(s)
Biomphalaria/parasitology , Disease Reservoirs , Schistosoma mansoni/isolation & purification , Schistosomiasis mansoni/veterinary , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rodent Diseases/parasitology , Rodent Diseases/transmission , Schistosoma mansoni/pathogenicity , Schistosomiasis mansoni/transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...