Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Am J Trop Med Hyg ; 107(2): 373-382, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35895421

ABSTRACT

This report summarizes the status of the global Dracunculiasis Eradication Program as of the end of 2021. Dracunculiasis (Guinea worm disease) has been eliminated from 17 of 21 countries where it was endemic in 1986, when an estimated 3.5 million cases occurred worldwide. Only Chad, Ethiopia, Mali, and South Sudan reported cases in humans in 2021. Chad, Ethiopia, and Mali also reported indigenous infections of animals, mostly domestic dogs, with Dracunculus medinensis. Insecurity and infections in animals are the main obstacles remaining to interrupting dracunculiasis transmission completely.


Subject(s)
Dracunculiasis , Humans , Animals , Dogs , Dracunculiasis/epidemiology , Dracunculiasis/prevention & control , Disease Eradication , Water Supply , Ethiopia/epidemiology , Chad/epidemiology
2.
Immunohorizons ; 6(6): 324-333, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35697476

ABSTRACT

Inhalation of ricin toxin (RT) elicits profuse inflammation and cell death within the upper and lower airways, ultimately culminating in acute respiratory distress syndrome. We previously reported that the effects of pulmonary RT exposure in mice are nullified by intranasal administration of an mAb mixture consisting of PB10, directed against ricin's enzymatic subunit (RTA), and SylH3, directed against ricin's binding subunit (RTB). We now report that delivery of PB10 and SylH3 as an RT-mAb immune complex (RIC) to mice by the intranasal or i.p. routes stimulates the rapid onset of RT-specific serum IgG that persists for months. RIC administration also induced high-titer, toxin-neutralizing Abs. Moreover, RIC-treated mice were immune to a subsequent 5 × LD50 RT challenge on days 30 or 90. Intranasal RIC administration was more effective than i.p. delivery at rendering mice immune to intranasal RT exposure. Finally, we found that the onset of RT-specific serum IgG following RIC delivery was independent of FcγR engagement, as revealed through FcγR knockout mice and RICs generated with PB10/SylH3 LALA (leucine to alanine) derivatives. In conclusion, a single dose of RICs given intranasally to mice was sufficient to stimulate durable protective immunity to RT by an FcγR-independent pathway.


Subject(s)
Ricin , Animals , Antibodies, Monoclonal , Antigen-Antibody Complex , Immunoglobulin G , Mice , Receptors, IgG , Ricin/chemistry , Ricin/metabolism
3.
Microorganisms ; 10(2)2022 Feb 01.
Article in English | MEDLINE | ID: mdl-35208785

ABSTRACT

The progression and systemic pathobiology of C. auris in the absence of a microbiota have not been described. Here, we describe the influence of the microbiota during the first 5 days of C. auris infection in germ-free or antibiotic-depleted mice. Depletion of the bacterial microbiota in both germ-free and antibiotic-depleted models results in a modest but important increase in the early stages of C. auris infection. Particularly the heart and lungs, followed by the cecum, uterus, and stomach, of intravenously (i.v.) infected neutropenic mice showed significant fungal organ burden. Understanding disease progression and pathobiology of C. auris in individuals with a depleted microbiota could potentially help in the development of care protocols that incorporate supplementation or restoration of the microbiota before invasive procedures, such as transplantation surgeries.

4.
Methods Mol Biol ; 2410: 305-324, 2022.
Article in English | MEDLINE | ID: mdl-34914054

ABSTRACT

Peyer's patches are organized gut-associated lymphoid tissues (GALT) in the small intestine and the primary route by which particulate antigens, including viruses and bacteria, are sampled by the mucosal immune system. Antigen sampling occurs through M cells, a specialized epithelial cell type located in the follicle-associated epithelium (FAE) that overlie Peyer's patch lymphoid follicles. While Peyer's patches play an integral role in intestinal homeostasis, they are also a gateway by which enteric pathogens, like Salmonella enterica serovar Typhimurium (STm), cross the intestinal barrier. Once pathogens like STm gain access to the underlying network of mucosal dendritic cells and macrophages they can spread systemically. Thus, Peyer's patches are at the crossroads of mucosal immunity and intestinal pathogenesis. In this chapter, we provide detailed methods to assess STm entry into mouse Peyer's patch tissues. We describe Peyer's patch collection methods and provide strategies to enumerate bacterial uptake. We also detail a method for quantifying bacterial shedding from infected animals and provide an immunohistochemistry protocol for the localization of STm along the gastrointestinal tract and insight into pathogen transit in the presence of protective antibodies. While the protocols are written for STm, they are easily tailored to other enteric pathogens.


Subject(s)
Salmonella typhimurium , Animals , Bacterial Vaccines , Immunity, Mucosal , Intestinal Mucosa , Mice , Peyer's Patches/immunology
5.
ACS Infect Dis ; 7(5): 1221-1235, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33728898

ABSTRACT

As the predominant antibody type in mucosal secretions, human colostrum, and breast milk, secretory IgA (SIgA) plays a central role in safeguarding the intestinal epithelium of newborns from invasive enteric pathogens like the Gram-negative bacterium Salmonella enterica serovar Typhimurium (STm). SIgA is a complex molecule, consisting of an assemblage of two or more IgA monomers, joining (J)-chain, and secretory component (SC), whose exact functions in neutralizing pathogens are only beginning to be elucidated. In this study, we produced and characterized a recombinant human SIgA variant of Sal4, a well-characterized monoclonal antibody (mAb) specific for the O5-antigen of STm lipopolysaccharide (LPS). We demonstrate by flow cytometry, light microscopy, and fluorescence microscopy that Sal4 SIgA promotes the formation of large, densely packed bacterial aggregates in vitro. In a mouse model, passive oral administration of Sal4 SIgA was sufficient to entrap STm within the intestinal lumen and reduce bacterial invasion into gut-associated lymphoid tissues by several orders of magnitude. Bacterial aggregates induced by Sal4 SIgA treatment in the intestinal lumen were recalcitrant to immunohistochemical staining, suggesting the bacteria were encased in a protective capsule. Indeed, a crystal violet staining assay demonstrated that STm secretes an extracellular matrix enriched in cellulose following even short exposures to Sal4 SIgA. Collectively, these results demonstrate that recombinant human SIgA recapitulates key biological activities associated with mucosal immunity and raises the prospect of oral passive immunization to combat enteric diseases.


Subject(s)
Immunoglobulin A, Secretory , Salmonella typhimurium , Agglutination , Humans , Immunity, Mucosal , Immunoglobulin A , Infant, Newborn , Intestinal Mucosa , Lymphoid Tissue
6.
NPJ Vaccines ; 5(1): 13, 2020.
Article in English | MEDLINE | ID: mdl-32128254

ABSTRACT

Inhalation of ricin toxin (RT), a Category B biothreat agent, provokes an acute respiratory distress syndrome marked by pro-inflammatory cytokine and chemokine production, neutrophilic exudate, and pulmonary edema. The severity of RT exposure is attributed to the tropism of the toxin's B subunit (RTB) for alveolar macrophages and airway epithelial cells, coupled with the extraordinarily potent ribosome-inactivating properties of the toxin's enzymatic subunit (RTA). While there are currently no vaccines or treatments approved to prevent RT intoxication, we recently described a humanized anti-RTA IgG1 MAb, huPB10, that was able to rescue non-human primates (NHPs) from lethal dose RT aerosol challenge if administered by intravenous (IV) infusion within hours of toxin exposure. We have now engineered an extended serum half-life variant of that MAb, huPB10-LS, and evaluated it as a pre-exposure prophylactic. Five Rhesus macaques that received a single intravenous infusion (25 mg/kg) of huPB10-LS survived a lethal dose aerosol RT challenge 28 days later, whereas three control animals succumbed to RT intoxication within 48 h. The huPB10-LS treated animals remained clinically normal in the hours and days following toxin insult, suggesting that pre-existing antibody levels were sufficient to neutralize RT locally. Moreover, pro-inflammatory markers in sera and BAL fluids collected 24 h following RT challenge were significantly dampened in huPB10-LS treated animals, as compared to controls. Finally, we found that all five surviving animals, within days after RT exposure, had anti-RT serum IgG titers against epitopes other than huPB10-LS, indicative of active immunization by residual RT and/or RT-immune complexes.

7.
Hum Vaccin Immunother ; 16(4): 793-807, 2020 04 02.
Article in English | MEDLINE | ID: mdl-31589555

ABSTRACT

Ricin toxin, a plant-derived, mannosylated glycoprotein, elicits an incapacitating and potentially lethal inflammatory response in the airways following inhalation. Uptake of ricin by alveolar macrophages (AM) and other pulmonary cell types occurs via two parallel pathways: one mediated by ricin's B subunit (RTB), a galactose-specific lectin, and one mediated by the mannose receptor (MR;CD206). Ricin's A subunit (RTA) is a ribosome-inactivating protein that triggers apoptosis in mammalian cells. It was recently reported that a single monoclonal antibody (MAb), PB10, directed against an immunodominant epitope on RTA and administered intravenously, was able to rescue Rhesus macaques from lethal aerosol dose of ricin. In this study, we now demonstrate in mice that the effectiveness PB10 is significantly improved when combined with a second MAb, SylH3, against RTB. Mice treated with PB10 alone survived lethal-dose intranasal ricin challenge, but experienced significant weight loss, moderate pulmonary inflammation (e.g., elevated IL-1 and IL-6 levels, PMN influx), and apoptosis of lung macrophages. In contrast, mice treated with the PB10/SylH3 cocktail were essentially impervious to pulmonary ricin toxin exposure, as evidenced by no weight loss, no change in local IL-1 and IL-6 levels, retention of lung macrophages, and a significant dampening of PMN recruitment into the bronchoalveolar lavage (BAL) fluids. The PB10/SylH3 cocktail only marginally reduced ricin binding to target cells in the BAL, suggesting that the antibody mixture neutralizes ricin by interfering with one or more steps in the RTB- and MR-dependent uptake pathways.


Subject(s)
Ricin , Animals , Antibodies, Monoclonal , Antibodies, Neutralizing , Inflammation/chemically induced , Lung , Macaca mulatta , Mice , Ricin/toxicity
8.
J Leukoc Biol ; 106(5): 1161-1176, 2019 11.
Article in English | MEDLINE | ID: mdl-31313388

ABSTRACT

Ricin toxin is a plant-derived, ribosome-inactivating protein that is rapidly cleared from circulation by Kupffer cells (KCs) and liver sinusoidal endothelial cells (LSECs)-with fatal consequences. Rather than being inactivated, ricin evades normal degradative pathways and kills both KCs and LSECs with remarkable efficiency. Uptake of ricin by these 2 specialized cell types in the liver occurs by 2 parallel routes: a "lactose-sensitive" pathway mediated by ricin's galactose/N-acetylgalactosamine-specific lectin subunit (RTB), and a "mannose-sensitive" pathway mediated by the mannose receptor (MR; CD206) or other C-type lectins capable of recognizing the mannose-side chains displayed on ricin's A (RTA) and B subunits. In this report, we investigated the capacity of a collection of ricin-specific mouse MAb and camelid single-domain (VH H) antibodies to protect KCs and LSECs from ricin-induced killing. In the case of KCs, individual MAbs against RTA or RTB afforded near complete protection against ricin in ex vivo and in vivo challenge studies. In contrast, individual MAbs or VH Hs afforded little (<40%) or even no protection to LSECs against ricin-induced death. Complete protection of LSECs was only achieved with MAb or VH H cocktails, with the most effective mixtures targeting RTA and RTB simultaneously. Although the exact mechanisms of protection of LSECs remain unknown, evidence indicates that the Ab cocktails exert their effects on the mannose-sensitive uptake pathway without the need for Fcγ receptor involvement. In addition to advancing our understanding of how toxins and small immune complexes are processed by KCs and LSECs, our study has important implications for the development of Ab-based therapies designed to prevent or treat ricin exposure should the toxin be weaponized.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/immunology , Antigen-Antibody Complex/toxicity , Endothelial Cells/immunology , Kupffer Cells/immunology , Liver/immunology , Ricin/toxicity , Animals , Antigen-Antibody Complex/immunology , Cell Line , Endothelial Cells/pathology , Female , Kupffer Cells/pathology , Liver/pathology , Mice , Ricin/immunology
9.
JCI Insight ; 4(1)2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30626745

ABSTRACT

Ricin toxin (RT) ranks at the top of the list of bioweapons of concern to civilian and military personnel alike, due to its high potential for morbidity and mortality after inhalation. In nonhuman primates, aerosolized ricin triggers severe acute respiratory distress characterized by perivascular and alveolar edema, neutrophilic infiltration, and severe necrotizing bronchiolitis and alveolitis. There are currently no approved countermeasures for ricin intoxication. Here, we report the therapeutic potential of a humanized mAb against an immunodominant epitope on ricin's enzymatic A chain (RTA). Rhesus macaques that received i.v. huPB10 4 hours after a lethal dose of ricin aerosol exposure survived toxin challenge, whereas control animals succumbed to ricin intoxication within 30 hours. Antibody intervention at 12 hours resulted in the survival of 1 of 5 monkeys. Changes in proinflammatory cytokine, chemokine, and growth factor profiles in bronchial alveolar lavage fluids before and after toxin challenge successfully clustered animals by treatment group and survival, indicating a relationship between local tissue damage and experimental outcome. This study represents the first demonstration, to our knowledge, in nonhuman primates that the lethal effects of inhalational ricin exposure can be negated by a drug candidate, and it opens up a path forward for product development.

10.
Am J Pathol ; 180(3): 1049-1058, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22222227

ABSTRACT

Macrophage differentiation and function are pivotal for cell survival from infection and involve the processing of microenvironmental signals that determine macrophage cell fate decisions to establish appropriate inflammatory balance. NADPH oxidase 2 (Nox2)-deficient chronic granulomatous disease (CGD) mice that lack the gp91(phox) (gp91(phox-/-)) catalytic subunit show high mortality rates compared with wild-type mice when challenged by infection with Listeria monocytogenes (Lm), whereas p47(phox)-deficient (p47(phox-/-)) CGD mice show survival rates that are similar to those of wild-type mice. We demonstrate that such survival results from a skewed macrophage differentiation program in p47(phox-/-) mice that favors the production of higher levels of alternatively activated macrophages (AAMacs) compared with levels of either wild-type or gp91(phox-/-) mice. Furthermore, the adoptive transfer of AAMacs from p47(phox-/-) mice can rescue gp91(phox-/-) mice during primary Lm infection. Key features of the protective function provided by p47(phox-/-) AAMacs against Lm infection are enhanced production of IL-1α and killing of Lm. Molecular analysis of this process indicates that p47(phox-/-) macrophages are hyperresponsive to IL-4 and show higher Stat6 phosphorylation levels and signaling coupled to downstream activation of AAMac transcripts in response to IL-4 stimulation. Notably, restoring p47(phox) protein expression levels reverts the p47(phox)-dependent AAMac phenotype. Our results indicate that p47(phox) is a previously unrecognized regulator for IL-4 signaling pathways that are important for macrophage cell fate choice.


Subject(s)
Cell Differentiation/physiology , Listeriosis/pathology , Macrophages/cytology , Membrane Glycoproteins/deficiency , NADPH Oxidases/deficiency , NADPH Oxidases/physiology , Signal Transduction/physiology , Adoptive Transfer , Animals , Genetic Vectors , Granulomatous Disease, Chronic/pathology , Interleukin-1alpha/biosynthesis , Interleukin-1alpha/pharmacology , Interleukin-4/pharmacology , Listeria monocytogenes , Macrophage Activation/physiology , Macrophages/physiology , Mice , Mice, Knockout , NADPH Oxidase 2 , Phagocytosis/physiology , Phosphorylation , STAT6 Transcription Factor/metabolism , Survival Analysis
11.
J Virol ; 85(1): 76-85, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20962087

ABSTRACT

While live attenuated influenza vaccines (LAIVs) have been shown to be efficacious and have been licensed for human use, the surface glycoproteins hemagglutinin (HA) and neuraminidase (NA) have to be updated for optimal protective efficacy. Little is known about the effect of different HA and NA proteins on the immunogenicity of LAIVs developed using the same backbone. A panel of LAIVs that share the internal protein genes, with unique HA and NA gene segments from different influenza subtypes, was rescued by reverse genetics, and a comparative study of immune responses induced by these vaccines was conducted in mice. The results suggest that the magnitude of lung immunity, including pulmonary IgA antibody and memory CD8(+) T lymphocytes, induced by the vaccines depends on the replication efficiency of the LAIVs, as well as the induction of cytokines/chemokines in the lungs. However, these factors are not important in determining systemic immunity such as serum antibody titers and memory CD8(+) T cells in the spleen. A qualitative analysis of immune responses induced by a single dose of an H5N1 LAIV revealed that the vaccine induced robust systemic and mucosal immunity in mice. In addition, antibodies and memory lymphocytes established in the lungs following vaccination were required for protection against lethal challenge with homologous and heterologous H5N1 viruses. Our results highlight the different requirements for inducing systemic and lung immunity that can be explored for the development of pulmonary immunity for protection against respiratory pathogens.


Subject(s)
Cytokines/biosynthesis , Influenza Vaccines/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Vaccines, Attenuated/immunology , Virus Replication , Animals , Antibodies, Viral/analysis , Antibodies, Viral/blood , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunoglobulin A/analysis , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Lung/chemistry , Mice , Mice, Inbred BALB C , Minute Virus of Mice , Neuraminidase/immunology , Orthomyxoviridae Infections/prevention & control , Vaccines, Attenuated/administration & dosage
12.
Proc Natl Acad Sci U S A ; 108(3): 1122-7, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21187399

ABSTRACT

Plasmodium falciparum has exerted tremendous selective pressure on genes that improve survival in severe malarial infections. Systemic lupus erythematosus (SLE) is an autoimmune disease that is six to eight times more prevalent in women of African descent than in women of European descent. Here we provide evidence that a genetic susceptibility to SLE protects against cerebral malaria. Mice that are prone to SLE because of a deficiency in FcγRIIB or overexpression of Toll-like receptor 7 are protected from death caused by cerebral malaria. Protection appears to be by immune mechanisms that allow SLE-prone mice better to control their overall inflammatory responses to parasite infections. These findings suggest that the high prevalence of SLE in women of African descent living outside of Africa may result from the inheritance of genes that are beneficial in the immune control of cerebral malaria but that, in the absence of malaria, contribute to autoimmune disease.


Subject(s)
Black People/genetics , Genetic Predisposition to Disease/genetics , Lupus Erythematosus, Systemic/genetics , Malaria, Cerebral/genetics , Plasmodium berghei/immunology , Receptors, IgG/deficiency , Toll-Like Receptor 7/metabolism , Animals , Brain/immunology , Brain/pathology , Cytokines/blood , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Erythrocytes/parasitology , Female , Flow Cytometry , Humans , Lupus Erythematosus, Systemic/ethnology , Malaria, Cerebral/immunology , Malaria, Cerebral/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Receptors, IgG/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spleen/physiology , Survival Analysis
13.
J Clin Invest ; 120(5): 1663-73, 2010 May.
Article in English | MEDLINE | ID: mdl-20389023

ABSTRACT

The target of neutralizing antibodies that protect against influenza virus infection is the viral protein HA. Genetic and antigenic variation in HA has been used to classify influenza viruses into subtypes (H1-H16). The neutralizing antibody response to influenza virus is thought to be specific for a few antigenically related isolates within a given subtype. However, while heterosubtypic antibodies capable of neutralizing multiple influenza virus subtypes have been recently isolated from phage display libraries, it is not known whether such antibodies are produced in the course of an immune response to influenza virus infection or vaccine. Here we report that, following vaccination with seasonal influenza vaccine containing H1 and H3 influenza virus subtypes, some individuals produce antibodies that cross-react with H5 HA. By immortalizing IgG-expressing B cells from 4 individuals, we isolated 20 heterosubtypic mAbs that bound and neutralized viruses belonging to several HA subtypes (H1, H2, H5, H6, and H9), including the pandemic A/California/07/09 H1N1 isolate. The mAbs used different VH genes and carried a high frequency of somatic mutations. With the exception of a mAb that bound to the HA globular head, all heterosubtypic mAbs bound to acid-sensitive epitopes in the HA stem region. Four mAbs were evaluated in vivo and protected mice from challenge with influenza viruses representative of different subtypes. These findings reveal that seasonal influenza vaccination can induce polyclonal heterosubtypic neutralizing antibodies that cross-react with the swine-origin pandemic H1N1 influenza virus and with the highly pathogenic H5N1 virus.


Subject(s)
Antibodies, Neutralizing/chemistry , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Animals , Antibodies, Monoclonal/chemistry , Binding Sites , Dogs , Epitope Mapping/methods , Female , Humans , Immunoglobulin G/metabolism , Leukocytes, Mononuclear/cytology , Mice , Mice, Inbred BALB C , Mutation , Orthomyxoviridae/metabolism , Protein Binding
14.
Am J Clin Pathol ; 120(6): 833-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14671971

ABSTRACT

The hemoparasite Babesia can cause life-threatening infections to neonates, elderly and immunocompromised people, and people who have undergone splenectomy. By using pooled hamster serum samples collected 21 days after infection with Babesia microti, we developed an immunohistochemical assay for formalin-fixed, paraffin-embedded tissue (FFPET) samples and blood smears. By use of the immunohistochemical assay, parasites were detected inside erythrocytes present in the heart, spleen, and liver of experimentally and naturally infected animals. FFPET samples from 2 fatal and 1 nonfatal human cases demonstrated immunohistochemical assay-positive parasites in circulating erythrocytes in various organs, including lymph nodes and spleen. In addition, air-dried blood smears from 4 patients showed positive immunohistochemical staining inside the erythrocytes. The immunohistochemical assay showed cross-reactivity against the Babesia WA-1 strain but did not react against Babesia bigemina or Plasmodium falciparum. The immunohistochemical assay for Babesia microti successfully detected parasites in human and animal FFPET samples and blood smears. This technique will be useful for the diagnosis of clinically suspected cases and for differentiating Babesia microti infection from malaria. Application of this technique to animal models will better define pathogenic mechanisms, including the possible recognition of exoerythrocytic tissue stages.


Subject(s)
Babesiosis/diagnosis , Immunohistochemistry/methods , Adult , Aged , Animals , Cross Reactions , Formaldehyde , Humans , Paraffin , Tissue Embedding
SELECTION OF CITATIONS
SEARCH DETAIL
...