Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Shock ; 44(6): 505-23, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26428845

ABSTRACT

Trauma is a leading cause of death in both military and civilian populations worldwide. Although medical advances have improved the overall morbidity and mortality often associated with trauma, additional research and innovative advancements in therapeutic interventions are needed to optimize patient outcomes. Cell-based therapies present a novel opportunity to improve trauma and critical care at both the acute and chronic phases that often follow injury. Although this field is still in its infancy, animal and human studies suggest that stem cells may hold great promise for the treatment of brain and spinal cord injuries, organ injuries, and extremity injuries such as those caused by orthopedic trauma, burns, and critical limb ischemia. However, barriers in the translation of cell therapies that include regulatory obstacles, challenges in manufacturing and clinical trial design, and a lack of funding are critical areas in need of development. In 2015, the Department of Defense Combat Casualty Care Research Program held a joint military-civilian meeting as part of its effort to inform the research community about this field and allow for effective planning and programmatic decisions regarding research and development. The objective of this article is to provide a "state of the science" review regarding cellular therapies in trauma and critical care, and to provide a foundation from which the potential of this emerging field can be harnessed to mitigate outcomes in critically ill trauma patients.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Critical Care/methods , Wounds and Injuries/therapy , Acute Kidney Injury/complications , Acute Kidney Injury/therapy , Animals , Blood Banks , Bone Marrow/physiopathology , Brain Injuries/therapy , Burns/complications , Burns/therapy , Cell- and Tissue-Based Therapy/standards , Clinical Trials as Topic , Compartment Syndromes/complications , Compartment Syndromes/therapy , Extremities/pathology , Humans , Ischemia/complications , Ischemia/therapy , Military Medicine , Orthopedics , Spinal Cord Injuries/therapy , Stem Cell Transplantation/legislation & jurisprudence , Stem Cell Transplantation/methods , Stem Cell Transplantation/standards , United States
2.
Pediatr Crit Care Med ; 14(4): 356-65, 2013 May.
Article in English | MEDLINE | ID: mdl-23548960

ABSTRACT

OBJECTIVE: Pertussis persists in the United States despite high immunization rates. This report characterizes the presentation and acute course of critical pertussis by quantifying demographic data, laboratory findings, clinical complications, and critical care therapies among children requiring admission to the PICU. DESIGN: Prospective cohort study. SETTING: Eight PICUs comprising the Eunice Kennedy Shriver National Institute for Child Health and Human Development Collaborative Pediatric Critical Care Research Network and 17 additional PICUs across the United States. PATIENTS: Eligible patients had laboratory confirmation of pertussis infection, were younger than 18 years old, and died in the PICU or were admitted to the PICU for at least 24 hours between June 2008 and August 2011. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: A total of 127 patients were identified. Median age was 49 days, and 105 (83%) patients were less than 3 months old. Fifty-five (43%) patients required mechanical ventilation and 12 patients (9.4%) died during initial hospitalization. Pulmonary hypertension was found in 16 patients (12.5%) and was present in 75% of patients who died, compared with 6% of survivors (p < 0.001). Median WBC was significantly higher in those requiring mechanical ventilation (p < 0.001), those with pulmonary hypertension (p < 0.001), and nonsurvivors (p < 0.001). Age, sex, and immunization status did not differ between survivors and nonsurvivors. Fourteen patients received leukoreduction therapy (exchange transfusion [12], leukopheresis [1], or both [1]). Survival benefit was not apparent. CONCLUSIONS: Pulmonary hypertension may be associated with mortality in pertussis critical illness. Elevated WBC is associated with the need for mechanical ventilation, pulmonary hypertension, and mortality risk. Research is indicated to elucidate how pulmonary hypertension, immune responsiveness, and elevated WBC contribute to morbidity and mortality and whether leukoreduction might be efficacious.


Subject(s)
Hypertension, Pulmonary/microbiology , Intensive Care Units, Pediatric/statistics & numerical data , Whooping Cough/complications , Whooping Cough/mortality , Bradycardia/microbiology , Exchange Transfusion, Whole Blood , Female , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/mortality , Infant , Leukapheresis , Leukocyte Count , Male , Pneumonia/microbiology , Premature Birth/epidemiology , Prospective Studies , Respiration, Artificial , Survival Rate , United States/epidemiology , Whooping Cough/blood , Whooping Cough/therapy
3.
Emerg Infect Dis ; 18(11): 1771-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23092514

ABSTRACT

Bordetella holmesii, a species closely related to B. pertussis, has been reported sporadically as a cause of whooping cough-like symptoms. To investigate whether B. pertussis-induced immunity is protective against infection with B. holmesii, we conducted an analysis using 11 human respiratory B. holmesii isolates collected during 2005-2009 from a highly B. pertussis-vaccinated population in Massachusetts. Neither whole-cell (wP) nor acellular (aP) B. pertussis vaccination conferred protection against these B. holmesii isolates in mice. Although T-cell responses induced by wP or aP cross-reacted with B. holmesii, vaccine-induced antibodies failed to efficiently bind B. holmesii. B. holmesii-specific antibodies provided in addition to wP were sufficient to rapidly reduce B. holmesii numbers in mouse lungs. Our findings suggest the established presence of B. holmesii in Massachusetts and that failure to induce cross-reactive antibodies may explain poor vaccine-induced cross-protection.


Subject(s)
Bordetella Infections/immunology , Bordetella pertussis/immunology , Bordetella/immunology , Cross Protection/immunology , Pertussis Vaccine/immunology , Adolescent , Adult , Animals , Antibodies, Bacterial/immunology , Antibody Specificity/immunology , Bordetella/classification , Bordetella/isolation & purification , Bordetella Infections/epidemiology , Child , Child, Preschool , Disease Susceptibility/immunology , Genes, Bacterial , Humans , Infant , Infant, Newborn , Massachusetts/epidemiology , Mice , Middle Aged , Phylogeny , Spleen/immunology , T-Lymphocytes/immunology , Whooping Cough/epidemiology , Whooping Cough/immunology , Whooping Cough/prevention & control , Young Adult
4.
Infect Immun ; 79(1): 527-41, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20974829

ABSTRACT

Interleukin-1 receptor-deficient (IL-1R(-/-)) mice are healthy despite being colonized by commensal microbes but are defective in defenses against specific pathogens, suggesting that IL-1R-mediated effects contribute to immune responses against specific pathogenic mechanisms. To better define the role of IL-1R in immunity to respiratory infections, we challenged IL-1R(-/-) mice with Bordetella pertussis and Bordetella parapertussis, the causative agents of whooping cough. Following inoculation with B. pertussis, but not B. parapertussis, IL-1R(-/-) mice showed elevated bacterial numbers and more extensive inflammatory pathology than wild-type mice. Acellular B. pertussis vaccines were not efficiently protective against B. pertussis in IL-1R(-/-) mice. B. pertussis-stimulated dendritic cells from IL-1R(-/-) mice produced higher levels of tumor necrosis factor alpha (TNF-α) and IL-6 than wild-type cells. Moreover, elevated levels of gamma interferon (IFN-γ) and TNF-α but lower levels of IL-10 were detected during B. pertussis infection in IL-1R(-/-) mice. Since B. parapertussis did not cause severe disease in IL-1R(-/-) mice, we hypothesized that the extreme requirement for IL-1R involves pertussis toxin (Ptx), which is expressed only by B. pertussis. An isogenic Ptx-deficient B. pertussis strain had only a modest phenotype in wild-type mice but was completely defective in causing lethal disease in IL-1R(-/-) mice, indicating that the particular virulence of B. pertussis in these mice requires Ptx. Ptx contributes to IL-1ß induction by B. pertussis, which is involved in IL-10 induction through IL-1R signaling. IL-10 treatment reduced B. pertussis numbers in IL-1R(-/-) mice, suggesting that the lower IL-10 responses partially account for the uncontrolled inflammation and bacterial growth in these mice.


Subject(s)
Bordetella pertussis/immunology , Pertussis Toxin/toxicity , Receptors, Interleukin-1 Type I/metabolism , Animals , Bordetella parapertussis/immunology , Cells, Cultured , Gene Expression Regulation , Interleukin-1/genetics , Interleukin-1/metabolism , Macrophages/physiology , Mice , Mice, Knockout , Receptors, Interleukin-1 Type I/genetics , Signal Transduction , Time
5.
Proc Biol Sci ; 277(1690): 2017-25, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20200027

ABSTRACT

Despite over 50 years of population-wide vaccination, whooping cough incidence is on the rise. Although Bordetella pertussis is considered the main causative agent of whooping cough in humans, Bordetella parapertussis infections are not uncommon. The widely used acellular whooping cough vaccines (aP) are comprised solely of B. pertussis antigens that hold little or no efficacy against B. parapertussis. Here, we ask how aP vaccination affects competitive interactions between Bordetella species within co-infected rodent hosts and thus the aP-driven strength and direction of in-host selection. We show that aP vaccination helped clear B. pertussis but resulted in an approximately 40-fold increase in B. parapertussis lung colony-forming units (CFUs). Such vaccine-mediated facilitation of B. parapertussis did not arise as a result of competitive release; B. parapertussis CFUs were higher in aP-relative to sham-vaccinated hosts regardless of whether infections were single or mixed. Further, we show that aP vaccination impedes host immunity against B. parapertussis-measured as reduced lung inflammatory and neutrophil responses. Thus, we conclude that aP vaccination interferes with the optimal clearance of B. parapertussis and enhances the performance of this pathogen. Our data raise the possibility that widespread aP vaccination can create hosts more susceptible to B. parapertussis infection.


Subject(s)
Bordetella Infections/microbiology , Bordetella parapertussis/pathogenicity , Diphtheria-Tetanus-acellular Pertussis Vaccines , Pertussis Vaccine , Whooping Cough/prevention & control , Animals , Bordetella Infections/complications , Bordetella Infections/epidemiology , Colony Count, Microbial , Diphtheria-Tetanus-acellular Pertussis Vaccines/administration & dosage , Disease Models, Animal , Female , Humans , Lung/microbiology , Mice , Mice, Inbred C57BL , Pertussis Vaccine/administration & dosage , Vaccination , Whooping Cough/complications , Whooping Cough/epidemiology , Whooping Cough/microbiology
6.
J Immunol ; 184(3): 1392-400, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20042578

ABSTRACT

Bordetella parapertussis causes the prolonged coughing illness known as pertussis or whooping cough, persisting for weeks within the respiratory tracts of infected hosts but inducing a very poor T cell response relative to that induced by Bordetella pertussis, the more common cause of pertussis. In this study, we examine the contributions of cytokines involved in the clearance of B. parapertussis and immunomodulation that delays effective clearance. The slow elimination of this pathogen from the respiratory tracts of mice coincides with the gradual accumulation of CD4(+) T cells in the lungs and B. parapertussis-responsive IFN-gamma-producing cells in the spleen. IFN-gamma-deficient mice were defective in the accumulation of leukocytes in lungs and in clearance of B. parapertussis from the lungs. In vitro B. parapertussis-stimulated macrophages produced IL-10, which inhibited the generation of the IFN-gamma response that is required for protection in vivo. As compared with wild-type mice, IL-10-deficient mice produced significantly higher levels of IFN-gamma, had higher numbers of leukocytes accumulated in the lungs, and cleared B. parapertussis more rapidly. Together, these data indicate that B. parapertussis induces the production of IL-10, which facilitates its persistence within infected hosts by limiting a protective IFN-gamma response.


Subject(s)
Bordetella parapertussis/immunology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Animals , Bordetella Infections/immunology , Bordetella Infections/microbiology , Bordetella Infections/pathology , Bordetella parapertussis/growth & development , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/pathology , Cell Line, Transformed , Cell Migration Inhibition/genetics , Cell Migration Inhibition/immunology , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interferon-gamma/deficiency , Interferon-gamma/physiology , Interleukin-10/deficiency , Interleukin-10/genetics , Interleukin-10/physiology , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
7.
Infect Immun ; 77(8): 3249-57, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19528223

ABSTRACT

Host immunity is a major driving force of antigenic diversity, resulting in pathogens that can evade immunity induced by closely related strains. Here we show that two Bordetella bronchiseptica strains, RB50 and 1289, express two antigenically distinct O-antigen serotypes (O1 and O2, respectively). When 18 additional B. bronchiseptica strains were serotyped, all were found to express either the O1 or O2 serotype. Comparative genomic hybridization and PCR screening showed that the expression of either the O1 or O2 serotype correlated with the strain containing either the classical or alternative O-antigen locus, respectively. Multilocus sequence typing analysis of 49 B. bronchiseptica strains was used to build a phylogenetic tree, which revealed that the two O-antigen loci did not associate with a particular lineage, evidence that these loci are horizontally transferred between B. bronchiseptica strains. From experiments using mice vaccinated with purified lipopolysaccharide from strain RB50 (O1), 1289 (O2), or RB50Deltawbm (O antigen deficient), our data indicate that these O antigens do not confer cross-protection in vivo. The lack of cross-immunity between O-antigen serotypes appears to contribute to inefficient antibody-mediated clearance between strains. Together, these data are consistent with the idea that the O-antigen loci of B. bronchiseptica are horizontally transferred between strains and encode antigenically distinct serotypes, resulting in inefficient cross-immunity.


Subject(s)
Bordetella Infections/microbiology , Bordetella bronchiseptica/genetics , Gene Transfer, Horizontal , O Antigens/genetics , Animals , Bacterial Typing Techniques , Bordetella Infections/immunology , Bordetella bronchiseptica/classification , Bordetella bronchiseptica/immunology , Bordetella bronchiseptica/isolation & purification , Cluster Analysis , Comparative Genomic Hybridization , DNA Fingerprinting , DNA, Bacterial/genetics , Genotype , Mice , Mice, Inbred C57BL , O Antigens/immunology , Phylogeny , Polymerase Chain Reaction , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...