Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 20(2): e1011992, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38416794

ABSTRACT

Recent advances in the in vitro cultivation of Cryptosporidium parvum using hollow fiber bioreactor technology (HFB) have permitted continuous growth of parasites that complete all life cycle stages. The method provides access to all stages of the parasite and provides a method for non-animal production of oocysts for use in clinical trials. Here we examined the effect of long-term (>20 months) in vitro culture on virulence-factors, genome conservation, and in vivo pathogenicity of the host by in vitro cultured parasites. We find low-level sequence variation that is consistent with that observed in calf-passaged parasites. Further using a calf model infection, oocysts obtained from the HFB caused diarrhea of the same volume, duration and oocyst shedding intensity as in vivo passaged parasites.


Subject(s)
Cryptosporidiosis , Cryptosporidium parvum , Cryptosporidium , Animals , Cryptosporidium parvum/genetics , Virulence , Cryptosporidiosis/parasitology , Oocysts , Genomics , Feces
2.
Antimicrob Agents Chemother ; 67(4): e0142522, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36920244

ABSTRACT

Recent advances on the development of bumped kinase inhibitors for treatment of cryptosporidiosis have focused on the 5-aminopyrazole-4-carboxamide scaffold, due to analogs that have less hERG inhibition, superior efficacy, and strong in vitro safety profiles. Three compounds, BKI-1770, -1841, and -1708, showed strong efficacy in C. parvum infected mice. Both BKI-1770 and BKI-1841 had efficacy in the C. parvum newborn calf model, reducing diarrhea and oocyst excretion. However, both compounds caused hyperflexion of the limbs seen as dropped pasterns. Toxicity experiments in rats and calves dosed with BKI-1770 showed enlargement of the epiphyseal growth plate at doses only slightly higher than the efficacious dose. Mice were used as a screen to check for bone toxicity, by changes to the tibia epiphyseal growth plate, or neurological causes, by use of a locomotor activity box. These results showed neurological effects from both BKI-1770 and BKI-1841 and bone toxicity in mice from BKI-1770, indicating one or both effects may be contributing to toxicity. However, BKI-1708 remains a viable treatment candidate for further evaluation as it showed no signs of bone toxicity or neurological effects in mice.


Subject(s)
Antineoplastic Agents , Antiprotozoal Agents , Cryptosporidiosis , Cryptosporidium parvum , Animals , Cattle , Mice , Rats , Cryptosporidiosis/drug therapy , Antiprotozoal Agents/pharmacology , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Oocysts
3.
Antimicrob Agents Chemother ; 66(1): e0156021, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34748385

ABSTRACT

Infection with Cryptosporidium spp. can cause severe diarrhea, leading to long-term adverse impacts and even death in malnourished children and immunocompromised patients. The only FDA-approved drug for treating cryptosporidiosis, nitazoxanide, has limited efficacy in the populations impacted the most by the diarrheal disease, and safe, effective treatment options are urgently needed. Initially identified by a large-scale phenotypic screening campaign, the antimycobacterial therapeutic clofazimine demonstrated great promise in both in vitro and in vivo preclinical models of Cryptosporidium infection. Unfortunately, a phase 2a clinical trial in HIV-infected adults with cryptosporidiosis did not identify any clofazimine treatment effect on Cryptosporidium infection burden or clinical outcomes. To explore whether clofazimine's lack of efficacy in the phase 2a trial may have been due to subtherapeutic clofazimine concentrations, a pharmacokinetic/pharmacodynamic modeling approach was undertaken to determine the relationship between clofazimine in vivo concentrations and treatment effects in multiple preclinical infection models. Exposure-response relationships were characterized using Emax and logistic models, which allowed predictions of efficacious clofazimine concentrations for the control and reduction of disease burden. After establishing exposure-response relationships for clofazimine treatment of Cryptosporidium infection in our preclinical model studies, it was unmistakable that the clofazimine levels observed in the phase 2a study participants were well below concentrations associated with anti-Cryptosporidium efficacy. Thus, despite a dosing regimen above the highest doses recommended for mycobacterial therapy, it is very likely the lack of treatment effect in the phase 2a trial was at least partially due to clofazimine concentrations below those required for efficacy against cryptosporidiosis. It is unlikely that clofazimine will provide a remedy for the large number of cryptosporidiosis patients currently without a viable treatment option unless alternative, safe clofazimine formulations with improved oral absorption are developed. (This study has been registered in ClinicalTrials.gov under identifier NCT03341767.).


Subject(s)
Antiprotozoal Agents , Cryptosporidiosis , Cryptosporidium , Adult , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Child , Clofazimine/pharmacology , Clofazimine/therapeutic use , Cryptosporidiosis/drug therapy , Diarrhea/drug therapy , Humans
4.
Vet Parasitol ; 289: 109336, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33418437

ABSTRACT

This is a review of the development of bumped-kinase inhibitors (BKIs) for the therapy of One Health parasitic apicomplexan diseases. Many apicomplexan infections are shared between humans and livestock, such as cryptosporidiosis and toxoplasmosis, as well as livestock only diseases such as neosporosis. We have demonstrated proof-of-concept for BKI therapy in livestock models of cryptosporidiosis (newborn calves infected with Cryptosporidium parvum), toxoplasmosis (pregnant sheep infected with Toxoplasma gondii), and neosporosis (pregnant sheep infected with Neospora caninum). We discuss the potential uses of BKIs for the treatment of diseases caused by apicomplexan parasites in animals and humans, and the improvements that need to be made to further develop BKIs.


Subject(s)
Antiparasitic Agents/pharmacology , Cryptosporidiosis/drug therapy , One Health , Piperidines/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology , Animals , Apicomplexa , Humans
5.
mBio ; 11(6)2020 12 15.
Article in English | MEDLINE | ID: mdl-33323514

ABSTRACT

The protozoan parasite Cryptosporidium sp. is a leading cause of diarrheal disease in those with compromised or underdeveloped immune systems, particularly infants and toddlers in resource-poor localities. As an enteric pathogen, Cryptosporidium sp. invades the apical surface of intestinal epithelial cells, where it resides in close proximity to metabolites in the intestinal lumen. However, the effect of gut metabolites on susceptibility to Cryptosporidium infection remains largely unstudied. Here, we first identified which gut metabolites are prevalent in neonatal mice when they are most susceptible to Cryptosporidium parvum infection and then tested the isolated effects of these metabolites on C. parvum invasion and growth in intestinal epithelial cells. Our findings demonstrate that medium or long-chain saturated fatty acids inhibit C. parvum growth, perhaps by negatively affecting the streamlined metabolism in C. parvum, which is unable to synthesize fatty acids. Conversely, long-chain unsaturated fatty acids enhanced C. parvum invasion, possibly by modulating membrane fluidity. Hence, gut metabolites, either from diet or produced by the microbiota, influence C. parvum growth in vitro and may also contribute to the early susceptibility to cryptosporidiosis seen in young animals.IMPORTANCECryptosporidium sp. occupies a unique intracellular niche that exposes the parasite to both host cell contents and the intestinal lumen, including metabolites from the diet and produced by the microbiota. Both dietary and microbial products change over the course of early development and could contribute to the changes seen in susceptibility to cryptosporidiosis in humans and mice. Consistent with this model, we show that the immature gut metabolome influenced the growth of Cryptosporidium parvumin vitro Interestingly, metabolites that significantly altered parasite growth were fatty acids, a class of molecules that Cryptosporidium sp. is unable to synthesize de novo The enhancing effects of polyunsaturated fatty acids and the inhibitory effects of saturated fatty acids presented in this study may provide a framework for future studies into this enteric parasite's interactions with exogenous fatty acids during the initial stages of infection.


Subject(s)
Bacteria/metabolism , Cryptosporidiosis/parasitology , Cryptosporidium parvum/physiology , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Intestinal Mucosa/parasitology , Animals , Animals, Newborn/metabolism , Animals, Newborn/microbiology , Animals, Newborn/parasitology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Cryptosporidiosis/metabolism , Cryptosporidiosis/microbiology , Cryptosporidium parvum/genetics , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/parasitology , Fatty Acids/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred ICR
6.
PLoS Pathog ; 16(5): e1008600, 2020 05.
Article in English | MEDLINE | ID: mdl-32453775

ABSTRACT

Apicomplexan parasites cause severe disease in both humans and their domesticated animals. Since these parasites readily develop drug resistance, development of new, effective drugs to treat infection caused by these parasites is an ongoing challenge for the medical and veterinary communities. We hypothesized that invertebrate-bacterial symbioses might be a rich source of anti-apicomplexan compounds because invertebrates are susceptible to infections with gregarines, parasites that are ancestral to all apicomplexans. We chose to explore the therapeutic potential of shipworm symbiotic bacteria as they are bona fide symbionts, are easily grown in axenic culture and have genomes rich in secondary metabolite loci [1,2]. Two strains of the shipworm symbiotic bacterium, Teredinibacter turnerae, were screened for activity against Toxoplasma gondii and one strain, T7901, exhibited activity against intracellular stages of the parasite. Bioassay-guided fractionation identified tartrolon E (trtE) as the source of the activity. TrtE has an EC50 of 3 nM against T. gondii, acts directly on the parasite itself and kills the parasites after two hours of treatment. TrtE exhibits nanomolar to picomolar level activity against Cryptosporidium, Plasmodium, Babesia, Theileria, and Sarcocystis; parasites representing all branches of the apicomplexan phylogenetic tree. The compound also proved effective against Cryptosporidium parvum infection in neonatal mice, indicating that trtE may be a potential lead compound for preclinical development. Identification of a promising new compound after such limited screening strongly encourages further mining of invertebrate symbionts for new anti-parasitic therapeutics.


Subject(s)
Antiprotozoal Agents , Apicomplexa/growth & development , Bivalvia/microbiology , Gammaproteobacteria/metabolism , Symbiosis , Animals , Antiprotozoal Agents/metabolism , Antiprotozoal Agents/pharmacology , Mice , Protozoan Infections/drug therapy
7.
Int J Parasitol ; 50(5): 413-422, 2020 05.
Article in English | MEDLINE | ID: mdl-32224121

ABSTRACT

Bumped Kinase Inhibitors, targeting Calcium-dependent Protein Kinase 1 in apicomplexan parasites with a glycine gatekeeper, are promising new therapeutics for apicomplexan diseases. Here we will review advances, as well as challenges and lessons learned regarding efficacy, safety, and pharmacology that have shaped our selection of pre-clinical candidates.


Subject(s)
Apicomplexa/drug effects , Coccidiosis/drug therapy , Protein Kinase Inhibitors , Animals , Apicomplexa/metabolism , Cryptosporidiosis/drug therapy , Cryptosporidium/drug effects , Cryptosporidium/metabolism , Humans , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/drug effects , Protein Kinases/metabolism , Toxoplasma/drug effects , Toxoplasma/metabolism , Toxoplasmosis/drug therapy
8.
Methods Mol Biol ; 2052: 253-282, 2020.
Article in English | MEDLINE | ID: mdl-31452167

ABSTRACT

Cryptosporidiosis, caused by the apicomplexan parasite Cryptosporidium parvum, is a moderate-to-severe diarrheal disease now recognized as one of the leading causes of morbidity and mortality in livestock globally, and in humans living in resource-limited parts of the world, particularly those with AIDS or malnourished individuals. This recognition has fueled efforts for the discovery of effective therapeutics. While recent progress in drug discovery has been encouraging, there are presently no acceptably effective parasite-specific drugs for the disease. The urgent need for new drug discovery or drug repurposing has also increased the need for refined animal models of clinical disease for therapeutic efficacy evaluation. Here, we describe an acute model of cryptosporidiosis using newborn calves to evaluate well-defined clinical and parasitological parameter outcomes, including the effect on diarrhea severity and duration, oocyst numbers produced, and multiple measures of clinical health. The model is highly reproducible and provides unequivocal direct measures of treatment efficacy on diarrhea severity and parasite replication.


Subject(s)
Cattle Diseases/drug therapy , Cryptosporidiosis/drug therapy , Cryptosporidium parvum/drug effects , Diarrhea/veterinary , Disease Models, Animal , Oocysts/drug effects , Animals , Cattle , Cattle Diseases/parasitology , Cattle Diseases/pathology , Cattle Diseases/urine , Cryptosporidiosis/parasitology , Cryptosporidiosis/pathology , Cryptosporidiosis/urine , Cryptosporidium parvum/growth & development , Cryptosporidium parvum/parasitology , Diarrhea/drug therapy , Diarrhea/parasitology , Diarrhea/pathology , Feces/parasitology , Humans , Infant, Newborn , Oocysts/growth & development , Oocysts/isolation & purification , Oocysts/metabolism , Workflow
9.
Nat Microbiol ; 3(7): 814-823, 2018 07.
Article in English | MEDLINE | ID: mdl-29946163

ABSTRACT

Stem-cell-derived organoids recapitulate in vivo physiology of their original tissues, representing valuable systems to model medical disorders such as infectious diseases. Cryptosporidium, a protozoan parasite, is a leading cause of diarrhoea and a major cause of child mortality worldwide. Drug development requires detailed knowledge of the pathophysiology of Cryptosporidium, but experimental approaches have been hindered by the lack of an optimal in vitro culture system. Here, we show that Cryptosporidium can infect epithelial organoids derived from human small intestine and lung. The parasite propagates within the organoids and completes its complex life cycle. Temporal analysis of the Cryptosporidium transcriptome during organoid infection reveals dynamic regulation of transcripts related to its life cycle. Our study presents organoids as a physiologically relevant in vitro model system to study Cryptosporidium infection.


Subject(s)
Cryptosporidiosis/genetics , Cryptosporidium/pathogenicity , Gene Expression Profiling/methods , Organoids/parasitology , Cryptosporidiosis/parasitology , Cryptosporidium/growth & development , Gene Expression Regulation , Humans , Intestine, Small/parasitology , Lung/parasitology , Models, Biological , Organ Culture Techniques , Sequence Analysis, RNA , Spatio-Temporal Analysis
10.
Am J Emerg Med ; 36(2): 341.e1-341.e3, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29074067

ABSTRACT

Allergic reaction is a common clinical picture in the Emergency Department (ED). Most allergic reactions are from food or drugs. A detailed history is an integral aspect of determining the causative agent of an allergy. Galactose-alpha-1,3-galactose (alpha-gal) allergy is a tick-acquired red meat allergy that causes delayed-onset allergic reaction or anaphylaxis due to molecular mimicry. Alpha-gal allergy may not be widely known as a cause of allergic reactions. Lack of universal awareness of this phenomenon in the ED and Urgent Care setting could lead to misdiagnosis, or delayed diagnosis. Subsequently, lack of proper instruction to avoid red meat could put patients at risk for future attacks with morbidity or mortality. We report three cases of allergic reaction presumed from red meat consumption secondary to alpha-gal allergy.


Subject(s)
Allergens/immunology , Disaccharides/immunology , Food Hypersensitivity/etiology , Red Meat/adverse effects , Tick Bites/complications , Ticks , Animals , Child , Female , Food Hypersensitivity/diagnosis , Food Hypersensitivity/immunology , Humans , Male , Middle Aged , Tick Bites/immunology
11.
Int J Parasitol ; 47(12): 753-763, 2017 10.
Article in English | MEDLINE | ID: mdl-28899690

ABSTRACT

Improvements have been made to the safety and efficacy of bumped kinase inhibitors, and they are advancing toward human and animal use for treatment of cryptosporidiosis. As the understanding of bumped kinase inhibitor pharmacodynamics for cryptosporidiosis therapy has increased, it has become clear that better compounds for efficacy do not necessarily require substantial systemic exposure. We now have a bumped kinase inhibitor with reduced systemic exposure, acceptable safety parameters, and efficacy in both the mouse and newborn calf models of cryptosporidiosis. Potential cardiotoxicity is the limiting safety parameter to monitor for this bumped kinase inhibitor. This compound is a promising pre-clinical lead for cryptosporidiosis therapy in animals and humans.


Subject(s)
Cryptosporidiosis/drug therapy , Cryptosporidium parvum/drug effects , Protein Kinase Inhibitors/therapeutic use , Administration, Oral , Animals , Animals, Newborn , Cattle , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Heart/drug effects , Humans , Inhibitory Concentration 50 , Interferon-gamma/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mutagenicity Tests , Pregnancy , Protein Binding , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/toxicity , Safety
12.
Article in English | MEDLINE | ID: mdl-28533246

ABSTRACT

Cryptosporidium parvum calcium-dependent protein kinase 1 (CpCDPK1) is a promising target for drug development against cryptosporidiosis. We report a series of low-nanomolar CpCDPK1 5-aminopyrazole-4-carboxamide (AC) scaffold inhibitors that also potently inhibit C. parvum growth in vitro Correlation between anti-CpCDPK1 and C. parvum growth inhibition, as previously reported for pyrazolopyrimidines, was not apparent. Nonetheless, lead AC compounds exhibited a substantial reduction of parasite burden in the neonatal mouse cryptosporidiosis model when dosed at 25 mg/kg.


Subject(s)
Antiprotozoal Agents/pharmacology , Cryptosporidiosis/drug therapy , Cryptosporidium parvum/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Animals , Antiprotozoal Agents/chemistry , Cryptosporidiosis/parasitology , Cryptosporidium parvum/growth & development , Mice , Protozoan Proteins/metabolism , Pyrazoles/chemistry , Pyrazoles/pharmacology
13.
J Infect Dis ; 216(1): 55-63, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28541457

ABSTRACT

There is a substantial need for novel therapeutics to combat the widespread impact caused by Crytosporidium infection. However, there is a lack of knowledge as to which drug pharmacokinetic (PK) characteristics are key to generate an in vivo response, specifically whether systemic drug exposure is crucial for in vivo efficacy. To identify which PK properties are correlated with in vivo efficacy, we generated physiologically based PK models to simulate systemic and gastrointestinal drug concentrations for a series of bumped kinase inhibitors (BKIs) that have nearly identical in vitro potency against Cryptosporidium but display divergent PK properties. When BKI concentrations were used to predict in vivo efficacy with a neonatal model of Cryptosporidium infection, these concentrations in the large intestine were the sole predictors of the observed in vivo efficacy. The significance of large intestinal BKI exposure for predicting in vivo efficacy was further supported with an adult mouse model of Cryptosporidium infection. This study suggests that drug exposure in the large intestine is essential for generating a superior in vivo response, and that physiologically based PK models can assist in the prioritization of leading preclinical drug candidates for in vivo testing.


Subject(s)
Cryptosporidiosis/drug therapy , Gastrointestinal Tract/drug effects , Protein Kinase Inhibitors/pharmacokinetics , Animals , Cryptosporidium parvum/drug effects , Cryptosporidium parvum/isolation & purification , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gastrointestinal Tract/metabolism , Inhibitory Concentration 50 , Mice , Mice, Knockout , Models, Theoretical , Naphthalenes/pharmacokinetics , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/blood , Pyrazoles/pharmacokinetics
14.
J Infect Dis ; 215(8): 1275-1284, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28329187

ABSTRACT

Bumped kinase inhibitors (BKIs) of Cryptosporidium parvum calcium-dependent protein kinase 1 (CpCDPK1) are leading candidates for treatment of cryptosporidiosis-associated diarrhea. Potential cardiotoxicity related to anti-human ether-à-go-go potassium channel (hERG) activity of the first-generation anti-Cryptosporidium BKIs triggered further testing for efficacy. A luminescence assay adapted for high-throughput screening was used to measure inhibitory activities of BKIs against C. parvum in vitro. Furthermore, neonatal and interferon γ knockout mouse models of C. parvum infection identified BKIs with in vivo activity. Additional iterative experiments for optimum dosing and selecting BKIs with minimum levels of hERG activity and frequencies of other safety liabilities included those that investigated mammalian cell cytotoxicity, C. parvum proliferation inhibition in vitro, anti-human Src inhibition, hERG activity, in vivo pharmacokinetic data, and efficacy in other mouse models. Findings of this study suggest that fecal concentrations greater than parasite inhibitory concentrations correlate best with effective therapy in the mouse model of cryptosporidiosis, but a more refined model for efficacy is needed.


Subject(s)
Antiprotozoal Agents/administration & dosage , Cryptosporidiosis/drug therapy , Cryptosporidium parvum/drug effects , Protein Kinase Inhibitors/administration & dosage , Administration, Oral , Animals , Diarrhea/drug therapy , Disease Models, Animal , Female , Mice , Mice, Knockout , Mice, SCID
15.
J Infect Dis ; 214(12): 1856-1864, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27923949

ABSTRACT

Cryptosporidiosis, caused by the apicomplexan parasite Cryptosporidium parvum, is a diarrheal disease that has produced a large global burden in mortality and morbidity in humans and livestock. There are currently no consistently effective parasite-specific pharmaceuticals available for this disease. Bumped kinase inhibitors (BKIs) specific for parasite calcium-dependent protein kinases (CDPKs) have been shown to reduce infection in several parasites having medical and veterinary importance, including Toxoplasma gondii, Plasmodium falciparum, and C. parvum In the present study, BKIs were screened for efficacy against C. parvum infection in the neonatal mouse model. Three BKIs were then selected for safety and clinical efficacy evaluation in the calf model for cryptosporidiosis. Significant BKI treatment effects were observed for virtually all clinical and parasitological scoring parameters, including diarrhea severity, oocyst shedding, and overall health. These results provide proof of concept for BKIs as therapeutic drug leads in an animal model for human cryptosporidiosis.


Subject(s)
Antiprotozoal Agents/administration & dosage , Cattle Diseases/drug therapy , Cryptosporidiosis/drug therapy , Protein Kinase Inhibitors/administration & dosage , Animals , Animals, Newborn , Antiprotozoal Agents/adverse effects , Cattle , Cryptosporidium parvum/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Mice, Inbred BALB C , Protein Kinase Inhibitors/adverse effects , Treatment Outcome
16.
PLoS Negl Trop Dis ; 7(7): e2307, 2013.
Article in English | MEDLINE | ID: mdl-23875042

ABSTRACT

Host defence peptides (HDPs) are expressed throughout the animal and plant kingdoms. They have multifunctional roles in the defence against infectious agents of mammals, possessing both bactericidal and immune-modulatory activities. We have identified a novel family of molecules secreted by helminth parasites (helminth defence molecules; HDMs) that exhibit similar structural and biochemical characteristics to the HDPs. Here, we have analyzed the functional activities of four HDMs derived from Schistosoma mansoni and Fasciola hepatica and compared them to human, mouse, bovine and sheep HDPs. Unlike the mammalian HDPs the helminth-derived HDMs show no antimicrobial activity and are non-cytotoxic to mammalian cells (macrophages and red blood cells). However, both the mammalian- and helminth-derived peptides suppress the activation of macrophages by microbial stimuli and alter the response of B cells to cytokine stimulation. Therefore, we hypothesise that HDMs represent a novel family of HDPs that evolved to regulate the immune responses of their mammalian hosts by retaining potent immune modulatory properties without causing deleterious cytotoxic effects.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Fasciola hepatica/immunology , Helminth Proteins/immunology , Host-Pathogen Interactions , Immunologic Factors/immunology , Macrophages/drug effects , Schistosoma mansoni/immunology , Animals , Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Bacteria/drug effects , Cattle , Cells, Cultured , Cytotoxins/metabolism , Erythrocytes/drug effects , Helminth Proteins/metabolism , Humans , Immunologic Factors/metabolism , Macrophage Activation/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Cathelicidins
17.
Vet Parasitol ; 188(1-2): 41-7, 2012 Aug 13.
Article in English | MEDLINE | ID: mdl-22455725

ABSTRACT

Cryptosporidium parvum is one of the main causes of diarrhea in neonatal calves resulting in significant morbidity and economic losses for producers worldwide. We have previously demonstrated efficacy of a new class of antimicrobial antibody fusions in a neonatal mouse model for C. parvum infection. Here, we extend efficacy testing of these products to experimental infection in calves, the principal target species. Neonatal calves were challenged with C. parvum oocysts and concomitantly treated with antibody-biocide fusion 4H9-G1-LL37 over the course of four days. This resulted in reduced severity of the disease when compared to control animals. Overall clinical health parameters showed significant improvement in treated animals. Oocyst shedding was reduced in treated when compared to control animals. Control of oocyst shedding is a prerequisite for breaking the cycle of re-infection on dairy farms. Antibody-biocide fusion products thus have the potential to reduce the impact of the infection in both individual animals and in the herd.


Subject(s)
Cattle Diseases/parasitology , Cryptosporidiosis/veterinary , Cryptosporidium parvum , Recombinant Proteins/therapeutic use , Animals , Antibodies, Protozoan/blood , Antibody Specificity , Cattle , Cattle Diseases/blood , Cryptosporidiosis/parasitology , Dairying , Feces/parasitology , Male , Mice , Oocysts , Parasite Egg Count , Recombinant Proteins/administration & dosage
18.
J Parasitol ; 98(1): 199-204, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21787211

ABSTRACT

The apicomplexan parasite Cryptosporidium parvum is an important cause of diarrhea in humans and cattle, and it can persistently infect immunocompromised hosts. No consistently effective parasite-specific pharmaceuticals or immunotherapies for control of cryptosporidiosis are presently available. The innate immune system represents the first line of host defense against a range of infectious agents, including parasitic protozoa. Several types of antimicrobial peptides and proteins, collectively referred to herein as biocides, constitute a major effector component of this system. In the present study, we evaluated lactoferrin, lactoferrin hydrolysate, 5 cationic peptides (lactoferricin B, cathelicidin LL37, indolicidin, ß-defensin 1, ß-defensin 2), lysozyme, and 2 phospholipases (phospholipase A2, and phosphatidylinositol-specific phospholipase C) for anti-cryptosporidial activity. The biocides were evaluated either alone or in combination with 3E2, a monoclonal antibody (MAb) against C. parvum that inhibits sporozoite attachment and invasion. Sporozoite viability and infectivity were used as indices of anti-cryptosporidial activity in vitro. All biocides except lactoferrin had a significant effect on sporozoite viability and infectivity. Lactoferrin hydrolysate and each of the 5 cationic peptides were highly parasiticidal and strongly reduced sporozoite infectivity. While each phospholipase also had parasiticidal activity, it was significantly less than that of lactoferrin hydrolysate and each of the cationic peptides. However, each phospholipase reduced sporozoite infectivity comparably to that observed with lactoferrin hydrolysate and the cationic peptides. Moreover, when 3 of the cationic peptides (cathelicidin LL37, ß-defensin 1, and ß-defensin 2) were individually combined with MAb 3E2, a significantly greater reduction of sporozoite infectivity was observed over that by 3E2 alone. In contrast, reduction of sporozoite infectivity by a combination of either phospholipase with MAb 3E2 was no greater than that by 3E2 alone. These collective observations suggest that cationic peptides and phospholipases neutralize C. parvum by mechanisms that are predominantly either parasiticidal or non-parasiticidal, respectively.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Cryptosporidiosis/drug therapy , Cryptosporidium parvum/drug effects , Phospholipases/pharmacology , Animals , Animals, Newborn , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antimicrobial Cationic Peptides/therapeutic use , Caco-2 Cells , Cattle , Cryptosporidium parvum/immunology , Cryptosporidium parvum/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Humans , Male , Mice , Mice, Inbred ICR , Phospholipases/therapeutic use , Specific Pathogen-Free Organisms , Sporozoites/drug effects , Sporozoites/physiology
19.
Antimicrob Agents Chemother ; 54(4): 1385-92, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20086143

ABSTRACT

At present no completely effective treatments are available for Cryptosporidium parvum infections in humans and livestock. Based on previous data showing the neutralizing potential of a panel of monoclonal antibodies developed against C. parvum, and based on the fact that innate immune peptides and enzymes have anticryptosporidial activity, we engineered several of these antibodies into antibody-biocide fusion proteins. We hypothesized that the combination of high-affinity antibody targeting with innate immune molecule-mediated killing would result in a highly effective new antiprotozoal agent. To test this hypothesis, we expressed antibody-biocide fusion proteins in a mammalian cell culture system and used the resulting products for in vitro and in vivo efficacy experiments. Antibody-biocide fusion proteins efficiently bound to, and destroyed, C. parvum sporozoites in vitro through a membrane-disruptive mechanism. When antibody-biocide fusion proteins were administered orally to neonatal mice in a prophylactic model of cryptosporidiosis, the induction of infection was reduced by as much as 81% in the mucosal epithelium of the gut, as determined on the basis of histopathological scoring of infectious stages. Several versions of antibody fusion proteins that differed in antigen specificity and in the biocide used had strong inhibitory effects on the initiation of infection. The results lay the groundwork for the development of a new class of antimicrobials effective against Cryptosporidium.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Protozoan/administration & dosage , Cryptosporidiosis/immunology , Cryptosporidiosis/prevention & control , Cryptosporidium parvum , Animals , Animals, Newborn , Antibodies, Monoclonal/genetics , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/genetics , Antibodies, Protozoan/genetics , Cryptosporidium parvum/immunology , Immunity, Innate , Mice , Mice, Inbred ICR , Molecular Sequence Data , Protein Engineering , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sporozoites/immunology
20.
Mol Biochem Parasitol ; 159(2): 138-41, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18400317

ABSTRACT

The biological basis for the specificity of host infectivity patterns of Cryptosporidium spp., in particular C. hominis and C. parvum, has yet to be fully elucidated. Comparison of the C. parvum and C. hominis P23 and GP900 predicted amino acid sequences revealed 3 differences in P23 and 4 and 17 differences in GP900 domains 1 and 5, respectively. Using monoclonal antibodies developed against the surface (glyco)proteins P23 and GP900 of the C. parvum Iowa isolate, solubilized glycoprotein from three C. hominis isolates was screened for reactivity using Western immunoblots. One of ten P23 MAbs and three of 21 GP900 MAbs were not reactive with any of the three C. hominis isolates. The non-reactive P23 MAb binds to a peptide epitope, while the non-reactive GP900 MAbs bind to either carbohydrate/carbohydrate-dependent or peptide epitopes of C. parvum. These results demonstrate phenotypic differences between C. hominis and C. parvum within two (glyco)proteins that are involved in parasite gliding motility and attachment/invasion.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Protozoan/metabolism , Antigens, Protozoan/immunology , Cryptosporidium/immunology , Protozoan Proteins/immunology , Amino Acid Substitution/genetics , Animals , Blotting, Western , Carbohydrates/immunology , DNA, Protozoan/chemistry , DNA, Protozoan/genetics , Epitopes/immunology , Humans , Membrane Glycoproteins/immunology , Membrane Proteins/immunology , Molecular Sequence Data , Protein Binding , Sequence Analysis, DNA , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...