Plasmodium chabaudi

Last updated

Plasmodium chabaudi
Scientific classification OOjs UI icon edit-ltr.svg
Domain: Eukaryota
Clade: Diaphoretickes
Clade: SAR
Clade: Alveolata
Phylum: Apicomplexa
Class: Aconoidasida
Order: Haemospororida
Family: Plasmodiidae
Genus: Plasmodium
Subgenus: Vinckeia
Species:
P. chabaudi
Binomial name
Plasmodium chabaudi
Landau, 1965
Subspecies

Plasmodium chabaudi is a parasite of the genus Plasmodium subgenus Vinckeia . As in all Plasmodium species, P. chabaudi has both vertebrate and insect hosts. The vertebrate hosts for this parasite are rodents. [1]

Contents

Taxonomy

This species was described in 1965 by Irène Landau. [2] It is named after the French parasitologist Alain Chabaud.

Subspecies

Two subspecies have been defined: P. chabaudi chabaudi and P. chabaudi adami. [3]

Genome

The nuclear genome is 18.8 megabases in size with a karyotype of 14 chromosomes. The G+C content is ~23%. A genome sequencing project is underway.

Distribution

P. chabaudi is found in Africa. It was first isolated from the blood of a shining thicket rat ( Thamnomys rutilans ) in the Central African Republic. [4]

Hosts

While it is difficult to study P. chabaudi in its natural host given the difficulty of taming the thicket rat, it has been studied extensively in laboratory mice, largely using the clone P. chabaudi chabaudi (AS). The pathology resembles that of human malaria in that animals are susceptible to parasite growth and pathology such as anemia, hypoglycemia, changes in body temperature, weight loss, and occasional death. The other cloned strains vary in growth rates and virulence. [5] One unique feature of this species is its prolonged course of infection. While it seems to persist for years in the thicket rat, P. chabaudi (AS) lasts up to three months in BALB/c or C57Bl/6 mice [6] P. falciparum has been observed to persist for up to a year, [7] and even in conditions of drought when there are no new infections. [8] Other species that are used to model human infection do not have this property. The other unique properties of this parasite are that it is synchronous, as first described for malaria by Galen, and that it prefers to infect normocytes, similar to P. falciparum , the most virulent human parasite, while several of the other rodent parasites have a preference for immature red blood cells, or reticulocytes, which they share with P. vivax.

In Anopheles stephensi the parasite synchronizes its circadian and diurnal rhythms with the host's. [9] Schneider et al., 2018 finds P. chabaudi is selected to take advantage of the cycles of feeding and lowered immunity of the mosquito. [9] They did not find any evidence for such a pattern in Mus musculus , testing for migration to peripheral vessels and finding none. [9] This parasite/mosquito synchronization is believed to hold for malaria parasites in general. [9]

Host resistance

Peak parasitaemia in Thamnomys rutilans the natural host is still unknown as of 2004. Landau 1965 and 66 did however find them to suffer to some severe degree, as did Ellerman 1940 in the sympatric and genetically close Grammomys surdaster . The peak is known to be 30% (109ml) for laboratory mice from many studies, including Jarra and Brown 1985. For specifically resistant breeds like C57Bl/6J Stevenson et al., 1982 finds the mortality is 5-20%, while for those known not to be resistant such as CBA/Ca and Dilute, Brown and non-Agouti (DBA), they find much higher mortalities. [10]

Lifecycle

There is usually a high female-to-male ratio in mature infections but this inhibits transmission at low densities due to lack of any male partner at the beginning of a new infection. [11] [12] [13] Therefore Reece et al., 2008 find P. chabaudi will bias toward a more even ratio at lower densities and when several clonal lineages are competing with each other in the same host. [11] [12] [13] This is believed to generalize beyond this species, to all Plasmodium. [11] [12] [13]

Therapeutic uses

P. chabaudi can reduce autoimmunity. Zinger et al., 2003 deliberately infected mice with the parasite and found reduced symptoms of autoimmunity. [14]

Related Research Articles

<span class="mw-page-title-main">Malaria</span> Mosquito-borne infectious disease

Malaria is a mosquito-borne infectious disease that affects humans and other vertebrates. Human malaria causes symptoms that typically include fever, fatigue, vomiting, and headaches. In severe cases, it can cause jaundice, seizures, coma, or death. Symptoms usually begin 10 to 15 days after being bitten by an infected Anopheles mosquito. If not properly treated, people may have recurrences of the disease months later. In those who have recently survived an infection, reinfection usually causes milder symptoms. This partial resistance disappears over months to years if the person has no continuing exposure to malaria.

<i>Plasmodium</i> Genus of parasitic protists that can cause malaria

Plasmodium is a genus of unicellular eukaryotes that are obligate parasites of vertebrates and insects. The life cycles of Plasmodium species involve development in a blood-feeding insect host which then injects parasites into a vertebrate host during a blood meal. Parasites grow within a vertebrate body tissue before entering the bloodstream to infect red blood cells. The ensuing destruction of host red blood cells can result in malaria. During this infection, some parasites are picked up by a blood-feeding insect, continuing the life cycle.

<i>Plasmodium falciparum</i> Protozoan species of malaria parasite

Plasmodium falciparum is a unicellular protozoan parasite of humans, and the deadliest species of Plasmodium that causes malaria in humans. The parasite is transmitted through the bite of a female Anopheles mosquito and causes the disease's most dangerous form, falciparum malaria. It is responsible for around 50% of all malaria cases. P. falciparum is therefore regarded as the deadliest parasite in humans. It is also associated with the development of blood cancer and is classified as a Group 2A (probable) carcinogen.

<i>Plasmodium malariae</i> Species of single-celled organism

Plasmodium malariae is a parasitic protozoan that causes malaria in humans. It is one of several species of Plasmodium parasites that infect other organisms as pathogens, also including Plasmodium falciparum and Plasmodium vivax, responsible for most malarial infection. Found worldwide, it causes a so-called "benign malaria", not nearly as dangerous as that produced by P. falciparum or P. vivax. The signs include fevers that recur at approximately three-day intervals – a quartan fever or quartan malaria – longer than the two-day (tertian) intervals of the other malarial parasite.

<i>Plasmodium gallinaceum</i> Bird malaria, including chicken

Plasmodium gallinaceum is a species of the genus Plasmodium that causes malaria in poultry.

<i>Plasmodium knowlesi</i> Species of single-celled organism

Plasmodium knowlesi is a parasite that causes malaria in humans and other primates. It is found throughout Southeast Asia, and is the most common cause of human malaria in Malaysia. Like other Plasmodium species, P. knowlesi has a life cycle that requires infection of both a mosquito and a warm-blooded host. While the natural warm-blooded hosts of P. knowlesi are likely various Old World monkeys, humans can be infected by P. knowlesi if they are fed upon by infected mosquitoes. P. knowlesi is a eukaryote in the phylum Apicomplexa, genus Plasmodium, and subgenus Plasmodium. It is most closely related to the human parasite Plasmodium vivax as well as other Plasmodium species that infect non-human primates.

<i>Plasmodium berghei</i> Single celled parasite, rodent malaria

Plasmodium berghei is a single-celled parasite causing rodent malaria. It is in the Plasmodium subgenus Vinckeia.

Plasmodium yoelii is a parasite of the genus Plasmodium subgenus Vinckeia. As in all Plasmodium species, P. yoelii has both vertebrate and insect hosts. The vertebrate hosts for this parasite are mammals.

<i>Laverania</i> Subgenus of single-celled organisms

Laverania is a subgenus of the parasite genus Plasmodium. Infection with these species results in malaria. The subgenus was first described in 1958.

<span class="mw-page-title-main">Malaria antigen detection tests</span>

Malaria antigen detection tests are a group of commercially available rapid diagnostic tests of the rapid antigen test type that allow quick diagnosis of malaria by people who are not otherwise skilled in traditional laboratory techniques for diagnosing malaria or in situations where such equipment is not available. There are currently over 20 such tests commercially available. The first malaria antigen suitable as target for such a test was a soluble glycolytic enzyme Glutamate dehydrogenase. None of the rapid tests are currently as sensitive as a thick blood film, nor as cheap. A major drawback in the use of all current dipstick methods is that the result is essentially qualitative. In many endemic areas of tropical Africa, however, the quantitative assessment of parasitaemia is important, as a large percentage of the population will test positive in any qualitative assay.

Vinckeia is a subgenus of the genus Plasmodium — all of which are parasitic alveolates. The subgenus Vinckeia was created by Cyril Garnham in 1964 to accommodate the mammalian parasites other than those infecting the primates.

Malaria vaccines are vaccines that prevent malaria, a mosquito-borne infectious disease which annually affects an estimated 247 million people worldwide and causes 619,000 deaths. The first approved vaccine for malaria is RTS,S, known by the brand name Mosquirix. As of April 2023, the vaccine has been given to 1.5 million children living in areas with moderate-to-high malaria transmission. It requires at least three doses in infants by age 2, and a fourth dose extends the protection for another 1–2 years. The vaccine reduces hospital admissions from severe malaria by around 30%.

<span class="mw-page-title-main">History of malaria</span> History of malaria infections

The history of malaria extends from its prehistoric origin as a zoonotic disease in the primates of Africa through to the 21st century. A widespread and potentially lethal human infectious disease, at its peak malaria infested every continent except Antarctica. Its prevention and treatment have been targeted in science and medicine for hundreds of years. Since the discovery of the Plasmodium parasites which cause it, research attention has focused on their biology as well as that of the mosquitoes which transmit the parasites.

Plasmodium vinckei is a parasite of the genus Plasmodium subgenus Vinckeia. As in all Plasmodium species, P. vinckei has both vertebrate and insect hosts. The vertebrate hosts for this parasite are rodents.

<span class="mw-page-title-main">Mass drug administration</span>

The administration of drugs to whole populations irrespective of disease status is referred to as mass drug administration (MDA) or mass dispensing.

Pregnancy-associated malaria (PAM) or placental malaria is a presentation of the common illness that is particularly life-threatening to both mother and developing fetus. PAM is caused primarily by infection with Plasmodium falciparum, the most dangerous of the four species of malaria-causing parasites that infect humans. During pregnancy, a woman faces a much higher risk of contracting malaria and of associated complications. Prevention and treatment of malaria are essential components of prenatal care in areas where the parasite is endemic – tropical and subtropical geographic areas. Placental malaria has also been demonstrated to occur in animal models, including in rodent and non-human primate models.

The mainstay of malaria diagnosis has been the microscopic examination of blood, utilizing blood films. Although blood is the sample most frequently used to make a diagnosis, both saliva and urine have been investigated as alternative, less invasive specimens. More recently, modern techniques utilizing antigen tests or polymerase chain reaction have been discovered, though these are not widely implemented in malaria endemic regions. Areas that cannot afford laboratory diagnostic tests often use only a history of subjective fever as the indication to treat for malaria.

Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) is a family of proteins present on the membrane surface of red blood cells that are infected by the malarial parasite Plasmodium falciparum. PfEMP1 is synthesized during the parasite's blood stage inside the RBC, during which the clinical symptoms of falciparum malaria are manifested. Acting as both an antigen and adhesion protein, it is thought to play a key role in the high level of virulence associated with P. falciparum. It was discovered in 1984 when it was reported that infected RBCs had unusually large-sized cell membrane proteins, and these proteins had antibody-binding (antigenic) properties. An elusive protein, its chemical structure and molecular properties were revealed only after a decade, in 1995. It is now established that there is not one but a large family of PfEMP1 proteins, genetically regulated (encoded) by a group of about 60 genes called var. Each P. falciparum is able to switch on and off specific var genes to produce a functionally different protein, thereby evading the host's immune system. RBCs carrying PfEMP1 on their surface stick to endothelial cells, which facilitates further binding with uninfected RBCs, ultimately helping the parasite to both spread to other RBCs as well as bringing about the fatal symptoms of P. falciparum malaria.

Irène Landau is a French parasitologist and professor emeritus at the National Museum of Natural History, France (MNHN) and Centre national de la recherche scientifique.

References

  1. Hoffman, Stephen P. (1996). Malaria Vaccine Development: A Multi-Immune Response Approach. American Society for Microbiology (ASM). ISBN   978-1-55581-111-2.
  2. Landau I (1965). "Description de Plasmodium chabaudi n. sp., parasite de rongeurs africains". C. R. Acad. Sci. 260: 3758–3761.
  3. Carter, R.; Walliker, D. (1976). "Malaria parasites of rodents of the Congo (Brazzaville): Plasmodium chabaudi adami subsp. nov. and Plasmodium vinckei lentum Landau, Michel, Adam and Boulard, 1970". Annales de Parasitologie Humaine et Comparée. 51 (6): 637–646. doi: 10.1051/parasite/1976516637 . ISSN   0003-4150. PMID   800328. Open Access logo PLoS transparent.svg
  4. Landau I, Killick-Kendrick R (1966). "Rodent plasmodia of the République Centrafricaine: the sporogony and tissue stages of Plasmodium chabaudi and P. berghei yoelii". Trans R Soc Trop Med Hyg. 60 (5): 633–49. doi:10.1016/0035-9203(66)90010-1. PMID   4163669.
  5. Stephens R, Culleton RL, Lamb TJ (Feb 2012). "The contribution of Plasmodium chabaudi to our understanding of malaria". Trends Parasitol. 28 (2): 73–82. doi:10.1016/j.pt.2011.10.006. PMC   4040349 . PMID   22100995.
  6. Achtman AH, Stephens R, Cadman ET, Harrison V, Langhorne J (Sep 2007). "Malaria-specific antibody responses and parasite persistence after infection of mice with Plasmodium chabaudi chabaudi". Parasite Immunol. 29 (9): 435–44. doi:10.1111/j.1365-3024.2007.00960.x. PMID   17727567. S2CID   24676716.
  7. Collins WE, Jeffery GM (May 2002). "A retrospective examination of sporozoite-induced and trophozoite-induced infections with Plasmodium ovale: development of parasitologic and clinical immunity during primary infection". Am J Trop Med Hyg. 66 (5): 492–502. doi: 10.4269/ajtmh.2002.66.492 . PMID   12201582.
  8. Characteristics (Oct 1998). "Plasmodium falciparum parasites that survive the lengthy dry season in eastern Sudan where malaria transmission is markedly seasonal. Babiker HA, Abdel-Muhsin AM, Ranford-Cartwright LC, Satti G, Walliker D". Am J Trop Med Hyg. 59 (4): 582–90. doi:10.4269/ajtmh.1998.59.582. PMID   9790434. S2CID   25489689.
  9. 1 2 3 4 Westwood, Mary L.; O'Donnell, Aidan J.; de Bekker, Charissa; Lively, Curtis M.; Zuk, Marlene; Reece, Sarah E. (2019-03-18). "The evolutionary ecology of circadian rhythms in infection" (PDF). Nature Ecology & Evolution . 3 (4). Nature Portfolio: 552–560. Bibcode:2019NatEE...3..552W. doi:10.1038/s41559-019-0831-4. hdl: 20.500.11820/6341ffdf-8682-4afe-a24d-0a82296900c3 . ISSN   2397-334X. PMID   30886375. S2CID   81985222.
  10. Mackinnon, Margaret J.; Read, Andrew F. (2004-06-29). "Virulence in malaria: an evolutionary viewpoint". Philosophical Transactions of the Royal Society B . 359 (1446). The Royal Society: 965–986. doi:10.1098/rstb.2003.1414. ISSN   0962-8436. PMC   1693375 . PMID   15306410.
  11. 1 2 3 Bousema, Teun; Drakeley, Chris (2011). "Epidemiology". Clinical Microbiology Reviews. 24 (2). American Society for Microbiology: 377–410. doi:10.1128/cmr.00051-10. ISSN   0893-8512. PMC   3122489 . PMID   21482730. S2CID   27743505.
  12. 1 2 3 Cornwallis, Charlie K.; Uller, Tobias (2010). "Towards an evolutionary ecology of sexual traits". Trends in Ecology & Evolution . 25 (3). Cell Press: 145–152. doi:10.1016/j.tree.2009.09.008. ISSN   0169-5347. PMID   19853321. S2CID   30059984.
  13. 1 2 3 Bousema, Teun; Okell, Lucy; Felger, Ingrid; Drakeley, Chris (2014-10-20). "Asymptomatic". Nature Reviews Microbiology . 12 (12). Nature Portfolio: 833–840. doi:10.1038/nrmicro3364. ISSN   1740-1526. PMID   25329408. S2CID   20524090.
  14. Shoenfeld, Yehuda (2004). "The idiotypic network in autoimmunity: antibodies that bind antibodies that bind antibodies". Nature . 10 (1). Nature Portfolio: 17–18. doi:10.1038/nm0104-17. ISSN   1078-8956. PMID   14702622. S2CID   3234263.