Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-32660993

ABSTRACT

Previously, ivermectin (1 to 10 mg/kg of body weight) was shown to inhibit the liver-stage development of Plasmodium berghei in orally dosed mice. Here, ivermectin showed inhibition of the in vitro development of Plasmodium cynomolgi schizonts (50% inhibitory concentration [IC50], 10.42 µM) and hypnozoites (IC50, 29.24 µM) in primary macaque hepatocytes when administered as a high dose prophylactically but not when administered in radical cure mode. The safety, pharmacokinetics, and efficacy of oral ivermectin (0.3, 0.6, and 1.2 mg/kg) with and without chloroquine (10 mg/kg) administered for 7 consecutive days were evaluated for prophylaxis or radical cure of P. cynomolgi liver stages in rhesus macaques. No inhibition or delay to blood-stage P. cynomolgi parasitemia was observed at any ivermectin dose (0.3, 0.6, and 1.2 mg/kg). Ivermectin (0.6 and 1.2 mg/kg) and chloroquine (10 mg/kg) in combination were well-tolerated with no adverse events and no significant pharmacokinetic drug-drug interactions observed. Repeated daily ivermectin administration for 7 days did not inhibit ivermectin bioavailability. It was recently demonstrated that both ivermectin and chloroquine inhibit replication of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in vitro Further ivermectin and chloroquine trials in humans are warranted to evaluate their role in Plasmodium vivax control and as adjunctive therapies against COVID-19 infections.


Subject(s)
Antimalarials/pharmacology , Chloroquine/pharmacology , Ivermectin/pharmacology , Liver/drug effects , Malaria/drug therapy , Plasmodium cynomolgi/drug effects , Animals , Antimalarials/blood , Antimalarials/pharmacokinetics , Biological Availability , Chloroquine/blood , Chloroquine/pharmacokinetics , Drug Administration Schedule , Drug Combinations , Drug Synergism , Female , Hepatocytes/drug effects , Hepatocytes/parasitology , Ivermectin/blood , Ivermectin/pharmacokinetics , Liver/parasitology , Macaca mulatta , Malaria/parasitology , Male , Parasitemia/drug therapy , Plasmodium cynomolgi/growth & development , Plasmodium cynomolgi/pathogenicity , Primary Cell Culture , Schizonts/drug effects , Schizonts/growth & development
2.
PLoS Pathog ; 15(9): e1007974, 2019 09.
Article in English | MEDLINE | ID: mdl-31536608

ABSTRACT

Plasmodium relapses are attributed to the activation of dormant liver-stage parasites and are responsible for a significant number of recurring malaria blood-stage infections. While characteristic of human infections caused by P. vivax and P. ovale, their relative contribution to malaria disease burden and transmission remains poorly understood. This is largely because it is difficult to identify 'bona fide' relapse infections due to ongoing transmission in most endemic areas. Here, we use the P. cynomolgi-rhesus macaque model of relapsing malaria to demonstrate that clinical immunity can form after a single sporozoite-initiated blood-stage infection and prevent illness during relapses and homologous reinfections. By integrating data from whole blood RNA-sequencing, flow cytometry, P. cynomolgi-specific ELISAs, and opsonic phagocytosis assays, we demonstrate that this immunity is associated with a rapid recall response by memory B cells that expand and produce anti-parasite IgG1 that can mediate parasite clearance of relapsing parasites. The reduction in parasitemia during relapses was mirrored by a reduction in the total number of circulating gametocytes, but importantly, the cumulative proportion of gametocytes increased during relapses. Overall, this study reveals that P. cynomolgi relapse infections can be clinically silent in macaques due to rapid memory B cell responses that help to clear asexual-stage parasites but still carry gametocytes.


Subject(s)
Immunity, Humoral , Malaria/immunology , Malaria/parasitology , Plasmodium cynomolgi/immunology , Plasmodium cynomolgi/pathogenicity , Animals , Antibodies, Protozoan/blood , B-Lymphocytes/immunology , Gene Expression Profiling , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Humans , Immunity, Humoral/genetics , Immunoglobulin G/blood , Immunologic Memory/genetics , Macaca mulatta , Malaria/genetics , Malaria, Vivax/genetics , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Male , Parasitemia/genetics , Parasitemia/immunology , Parasitemia/parasitology , Plasmodium vivax/immunology , Plasmodium vivax/pathogenicity , Recurrence , Sporozoites/immunology , Sporozoites/pathogenicity
3.
Am J Trop Med Hyg ; 97(2): 548-555, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28829738

ABSTRACT

Histopathological data collected from patients with severe malaria have been instrumental for studying malaria pathogenesis. Animal models of malaria are critical to complement such studies. Here, the histopathological changes observed in a rhesus macaque with severe and complicated Plasmodium cynomolgi malaria are reported. The animal presented with thrombocytopenia, severe anemia, and hyperparasitemia during the acute infection. The macaque was given subcurative antimalarial treatment, fluid support, and a blood transfusion to treat the clinical complications, but at the time of transfusion, kidney function was compromised. These interventions did not restore kidney function, and the animal was euthanized due to irreversible renal failure. Gross pathological and histological examinations revealed that the lungs, kidneys, liver, spleen, and bone marrow exhibited abnormalities similar to those described in patients with malaria. Overall, this case report illustrates the similarities in the pathophysiological complications that can occur in human malaria and cynomolgi malaria in rhesus macaques.


Subject(s)
Macaca mulatta/parasitology , Malaria/complications , Malaria/parasitology , Plasmodium cynomolgi/isolation & purification , Plasmodium cynomolgi/parasitology , Plasmodium cynomolgi/pathogenicity , Animals , Bone Marrow/anatomy & histology , Disease Models, Animal , Humans , Kidney/cytology , Liver/cytology , Lung/cytology , Malaria/pathology , Spleen/cytology
4.
Parasitol Int ; 64(3): 274-81, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25316604

ABSTRACT

Although the human malaria parasite Plasmodium vivax is closely related to Asian Old World monkey malaria parasites, there are no reports of P. vivax infections in macaques. In this study, we compared the infectivity of P. vivax and Plasmodium cynomolgi in Japanese macaques (Macaca fuscata) and in cynomolgus macaques (Macaca fascicularis). The Japanese macaques were highly susceptible to P. cynomolgi but not to P. vivax, whereas cynomolgus macaques showed mild/limited P. cynomolgi infection and were, also, not susceptible to P. vivax. Serotyping and amino acid sequence comparison of erythrocyte surface Duffy antigen/receptor for chemokines (DARC) indicate that the Japanese macaque DARC sequence is nearly identical to that of rhesus (Macaca mulatta) and cynomolgus macaques. This suggests that the macaques share a common mechanism for preventing P. vivax infection. Comparison of amino acid sequences of the Duffy-binding-like (DBL) domain from several different Plasmodium species suggests that P. vivax DBLs will not bind to macaque DARCs, which can explain the lack of P. vivax infectivity. The DBL sequence analyses also suggest that P. cynomolgi DBLs may target Japanese macaque erythrocytes through a DARC-independent interaction.


Subject(s)
Antigens, Protozoan/genetics , Macaca/parasitology , Malaria, Vivax/veterinary , Malaria/veterinary , Monkey Diseases/parasitology , Plasmodium cynomolgi/pathogenicity , Plasmodium vivax/pathogenicity , Protozoan Proteins/genetics , Receptors, Cell Surface/genetics , Amino Acid Sequence , Animals , Antigens, Protozoan/chemistry , Disease Susceptibility , Humans , Macaca/blood , Macaca/genetics , Macaca/immunology , Macaca fascicularis/blood , Macaca fascicularis/genetics , Macaca fascicularis/immunology , Macaca mulatta/blood , Macaca mulatta/genetics , Macaca mulatta/immunology , Malaria/parasitology , Malaria, Vivax/parasitology , Molecular Sequence Data , Plasmodium cynomolgi/genetics , Plasmodium vivax/genetics , Protozoan Proteins/chemistry , Receptors, Cell Surface/chemistry , Serotyping , Species Specificity
5.
Am J Trop Med Hyg ; 86(6): 931-5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22665596

ABSTRACT

There remains a need for new drugs to prevent relapse of Plasmodium vivax or P. ovale infection. The relapsing primate malaria P. cynomolgi has been used for decades to assess drugs for anti-hypnozoite activity. After sporozoite inoculation and blood-stage cure of initial parasitemia with chloroquine, rhesus macaques were treated on subsequent relapses with chloroquine in conjunction with test regimens of approved drugs. Tested drugs were selected for known liver or blood-stage activity and were tested alone or in conjunction with low-dose primaquine. Tinidazole and pyrazinamide prevented relapse when used in conjunction with chloroquine and low-dose primaquine. Triamterene and tinidazole administered without primaquine achieved radical cure in some animals. All other tested drugs or combinations failed to prevent relapse. The rhesus macaque-P. cynomolgi model remains a useful tool for screening drugs with anti-hypnozoite activity. Tinidazole and pyrazinamide require further investigation as agents to enable dose reduction of primaquine.


Subject(s)
Antimalarials/therapeutic use , Macaca mulatta/parasitology , Malaria, Vivax/drug therapy , Plasmodium cynomolgi/drug effects , Plasmodium cynomolgi/pathogenicity , Animals , Chloroquine/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Malaria, Vivax/prevention & control , Parasitemia/prevention & control , Plasmodium vivax/growth & development , Plasmodium vivax/pathogenicity , Primaquine/therapeutic use , Pyrazinamide/therapeutic use , Secondary Prevention , Sporozoites/drug effects , Tinidazole/therapeutic use , Triamterene/therapeutic use
6.
Infect Immun ; 78(3): 1032-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20048045

ABSTRACT

Both Plasmodium and Babesia species are intraerythrocytic protozoans that infect a wide range of hosts, including humans, and they elicit similar inflammatory responses and clinical manifestations that differ markedly in severity. We recently reported that a rhesus macaque that was chronically infected with Babesia microti was able to control infection with Plasmodium cynomolgi (a parasite of macaques with characteristics very similar to those of Plasmodium vivax) better than naïve monkeys. To confirm this and to investigate the underlying immunopathology, six naïve rhesus monkeys were infected with B. microti. After 24 days, four of these monkeys and four naïve rhesus monkeys were challenged with P. cynomolgi blood-stage parasites. B. microti persisted at low levels in all monkeys, and the clinical parameters were comparable to those of noninfected controls. There was a significant decrease in P. cynomolgi parasitemia in animals coinfected with B. microti compared to the parasitemia in animals infected with P. cynomolgi alone. This decrease in P. cynomolgi parasitemia correlated with increases in the levels of proinflammatory monocytes at the time of P. cynomolgi infection and with higher C-reactive protein (CRP) serum levels 1 week after malaria infection. Therefore, we conclude that ongoing infection with B. microti parasites leads to suppression of malaria infection.


Subject(s)
Babesia microti/pathogenicity , Babesiosis/complications , Babesiosis/pathology , Malaria/complications , Malaria/pathology , Monkey Diseases/parasitology , Plasmodium cynomolgi/pathogenicity , Animals , Babesiosis/parasitology , Blood/immunology , Blood/parasitology , Blood Chemical Analysis , C-Reactive Protein/analysis , Disease Models, Animal , Female , Humans , Macaca mulatta , Malaria/parasitology , Monocytes/immunology , Parasitemia
7.
J Vector Borne Dis ; 44(2): 116-21, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17722865

ABSTRACT

BACKGROUND & OBJECTIVES: Early gestational malaria is more deleterious than late gestational infection. Still the pathophysiology of maternofoetal organ--the placenta in malaria remains almost unexplored during early gestation. Present study dealing with oxidoreductases in early gestational placenta during maternal malarial infection of Plasmodium cynomolgi bastianellii in rhesus monkeys was anticipated to provide a better insight into the functional impairment of this organ leading to foetal abnormalities. METHODS: Three control and four experimental monkeys (Macaca mulatta) were quarantined for one month prior to experimentation. Experimental monkeys at 2- 2 1/2 months of gestation were inoculated with P. cynomolgi bastianellii. On attaining first peak of parasitaemia the placentae were collected from anesthetised animals. The snap-frozen, cryostat sections were subjected to histochemical localisation for 3 (or 17) beta-hydroxysteroid dehydrogenase (beta-HSD) [3 (or 17) beta-hydroxysteroid: NAD (P+) oxidoreductase, EC 1.1.1.51 hydroxysteroid dehydrogenases] and NADPH-tetrazolium reductase [NADPH: (acceptor) oxidoreductase, EC 1.6.99.1 NADPH-TR]. Comparative microscopy of control and malaria infected placental sections was performed and analysed. RESULTS: A localised decrease in both the enzymes was observed in syncytiotrophoblast layer of malaria infected monkey placenta. The areas showing morphological damage of syncytiotrophoblast were also depicting gross reduction in NADPH-TR activity. INTERPRETATION & CONCLUSION: The altered enzymatic activities [3 (or 17) beta-HSD and NADPH-TR] in malaria infected early gestational monkey placenta have been discussed in the light of placental function. It could be concluded by present studies that these alterations would affect the cellular metabolism especially steroidogenesis and detoxification process which in turn would affect the normal development of the foetus as well as maintenance of gestation.


Subject(s)
Malaria/enzymology , Oxidoreductases/metabolism , Placenta/enzymology , Plasmodium cynomolgi/pathogenicity , Pregnancy Complications, Parasitic/enzymology , Animals , Disease Models, Animal , Female , Macaca mulatta , Malaria/parasitology , Placenta/pathology , Pregnancy
8.
J Parasitol ; 92(1): 202-5, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16629341

ABSTRACT

Sporozoites of 3 isolates of Plasmodium cynomolgi dissected from the salivary glands of Anopheles dirus and Anopheles quadrimaculatus were injected intravenously into 9 New World monkeys. Liver stage parasites were demonstrated in all 9 animals; 7 of these animals also produced blood stages after prepatent periods of 9 to 23 days.


Subject(s)
Aotidae/parasitology , Hepatocytes/parasitology , Malaria/veterinary , Plasmodium cynomolgi/pathogenicity , Saimiri/parasitology , Animals , Anopheles/parasitology , Erythrocytes/parasitology , Injections, Intravenous/veterinary , Macaca mulatta , Malaria/parasitology , Malaria/pathology , Malaria/transmission , Plasmodium cynomolgi/isolation & purification , Plasmodium cynomolgi/physiology , Sporozoites/pathogenicity , Time Factors
10.
J Infect Dis ; 191(3): 400-9, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15633100

ABSTRACT

We used human microarrays to examine gene expression in a rhesus monkey model of human Plasmodium vivax malaria (P. cynomolgi in Macaca mulatta). Whole-blood cells were collected for extraction of RNA before infection, during both the initial liver phase of infection and bloodstream infection, and during the course of 2 bloodstream relapses. Clustering analysis showed that similarities in gene expression were greater at similar stages of the protocol for the 2 different monkeys than for the same monkey at different stages of the protocol. Interestingly, a large number of genes involved in RNA processing showed distinct down-regulation during the initial liver phase of infection. When only up-regulated genes were examined, there was evidence of an increasing number of "defense response" genes as the infection evolved but not of "cytoskeleton" genes (P

Subject(s)
Disease Models, Animal , Malaria/metabolism , Oligonucleotide Array Sequence Analysis/methods , Plasmodium cynomolgi/pathogenicity , Proteins/metabolism , Proteome , Transcription, Genetic , Animals , Gene Expression Profiling , Gene Expression Regulation , Humans , Macaca mulatta , Malaria/genetics , Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...