Overexpression of <named-content content-type="genus-species">Plasmodium berghei</named-content> ATG8 by Liver Forms Leads to Cumulative Defects in Organelle Dynamics and to Generation of Noninfectious Merozoites

ABSTRACT Plasmodium parasites undergo continuous cellular renovation to adapt to various environments in the vertebrate host and insect vector. In hepatocytes, Plasmodium berghei discards unneeded organelles for replication, such as micronemes involved in invasion. Concomitantly, intrahepatic parasi...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Christiane Voss, Karen Ehrenman, Godfree Mlambo, Satish Mishra, Kota Arun Kumar, John B. Sacci, Photini Sinnis, Isabelle Coppens
Formato: article
Lenguaje:EN
Publicado: American Society for Microbiology 2016
Materias:
Acceso en línea:https://doaj.org/article/9f9ba6d42b60419d9b7eb61fbe9c7b10
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:9f9ba6d42b60419d9b7eb61fbe9c7b10
record_format dspace
spelling oai:doaj.org-article:9f9ba6d42b60419d9b7eb61fbe9c7b102021-11-15T15:50:16ZOverexpression of <named-content content-type="genus-species">Plasmodium berghei</named-content> ATG8 by Liver Forms Leads to Cumulative Defects in Organelle Dynamics and to Generation of Noninfectious Merozoites10.1128/mBio.00682-162150-7511https://doaj.org/article/9f9ba6d42b60419d9b7eb61fbe9c7b102016-07-01T00:00:00Zhttps://journals.asm.org/doi/10.1128/mBio.00682-16https://doaj.org/toc/2150-7511ABSTRACT Plasmodium parasites undergo continuous cellular renovation to adapt to various environments in the vertebrate host and insect vector. In hepatocytes, Plasmodium berghei discards unneeded organelles for replication, such as micronemes involved in invasion. Concomitantly, intrahepatic parasites expand organelles such as the apicoplast that produce essential metabolites. We previously showed that the ATG8 conjugation system is upregulated in P. berghei liver forms and that P. berghei ATG8 (PbATG8) localizes to the membranes of the apicoplast and cytoplasmic vesicles. Here, we focus on the contribution of PbATG8 to the organellar changes that occur in intrahepatic parasites. We illustrated that micronemes colocalize with PbATG8-containing structures before expulsion from the parasite. Interference with PbATG8 function by overexpression results in poor development into late liver stages and production of small merosomes that contain immature merozoites unable to initiate a blood infection. At the cellular level, PbATG8-overexpressing P. berghei exhibits a delay in microneme compartmentalization into PbATG8-containing autophagosomes and elimination compared to parasites from the parental strain. The apicoplast, identifiable by immunostaining of the acyl carrier protein (ACP), undergoes an abnormally fast proliferation in mutant parasites. Over time, the ACP staining becomes diffuse in merosomes, indicating a collapse of the apicoplast. PbATG8 is not incorporated into the progeny of mutant parasites, in contrast to parental merozoites in which PbATG8 and ACP localize to the apicoplast. These observations reveal that Plasmodium ATG8 is a key effector in the development of merozoites by controlling microneme clearance and apicoplast proliferation and that dysregulation in ATG8 levels is detrimental for malaria infectivity. IMPORTANCE Malaria is responsible for more mortality than any other parasitic disease. Resistance to antimalarial medicines is a recurring problem; new drugs are urgently needed. A key to the parasite’s successful intracellular development in the liver is the metabolic changes necessary to convert the parasite from a sporozoite to a replication-competent, metabolically active trophozoite form. Our study reinforces the burgeoning concept that organellar changes during parasite differentiation are mediated by an autophagy-like process. We have identified ATG8 in Plasmodium liver forms as an important effector that controls the development and fate of organelles, e.g., the clearance of micronemes that are required for hepatocyte invasion and the expansion of the apicoplast that produces many metabolites indispensable for parasite replication. Given the unconventional properties and the importance of ATG8 for parasite development in hepatocytes, targeting the parasite’s autophagic pathway may represent a novel approach to control malarial infections.Christiane VossKaren EhrenmanGodfree MlamboSatish MishraKota Arun KumarJohn B. SacciPhotini SinnisIsabelle CoppensAmerican Society for MicrobiologyarticleMicrobiologyQR1-502ENmBio, Vol 7, Iss 3 (2016)
institution DOAJ
collection DOAJ
language EN
topic Microbiology
QR1-502
spellingShingle Microbiology
QR1-502
Christiane Voss
Karen Ehrenman
Godfree Mlambo
Satish Mishra
Kota Arun Kumar
John B. Sacci
Photini Sinnis
Isabelle Coppens
Overexpression of <named-content content-type="genus-species">Plasmodium berghei</named-content> ATG8 by Liver Forms Leads to Cumulative Defects in Organelle Dynamics and to Generation of Noninfectious Merozoites
description ABSTRACT Plasmodium parasites undergo continuous cellular renovation to adapt to various environments in the vertebrate host and insect vector. In hepatocytes, Plasmodium berghei discards unneeded organelles for replication, such as micronemes involved in invasion. Concomitantly, intrahepatic parasites expand organelles such as the apicoplast that produce essential metabolites. We previously showed that the ATG8 conjugation system is upregulated in P. berghei liver forms and that P. berghei ATG8 (PbATG8) localizes to the membranes of the apicoplast and cytoplasmic vesicles. Here, we focus on the contribution of PbATG8 to the organellar changes that occur in intrahepatic parasites. We illustrated that micronemes colocalize with PbATG8-containing structures before expulsion from the parasite. Interference with PbATG8 function by overexpression results in poor development into late liver stages and production of small merosomes that contain immature merozoites unable to initiate a blood infection. At the cellular level, PbATG8-overexpressing P. berghei exhibits a delay in microneme compartmentalization into PbATG8-containing autophagosomes and elimination compared to parasites from the parental strain. The apicoplast, identifiable by immunostaining of the acyl carrier protein (ACP), undergoes an abnormally fast proliferation in mutant parasites. Over time, the ACP staining becomes diffuse in merosomes, indicating a collapse of the apicoplast. PbATG8 is not incorporated into the progeny of mutant parasites, in contrast to parental merozoites in which PbATG8 and ACP localize to the apicoplast. These observations reveal that Plasmodium ATG8 is a key effector in the development of merozoites by controlling microneme clearance and apicoplast proliferation and that dysregulation in ATG8 levels is detrimental for malaria infectivity. IMPORTANCE Malaria is responsible for more mortality than any other parasitic disease. Resistance to antimalarial medicines is a recurring problem; new drugs are urgently needed. A key to the parasite’s successful intracellular development in the liver is the metabolic changes necessary to convert the parasite from a sporozoite to a replication-competent, metabolically active trophozoite form. Our study reinforces the burgeoning concept that organellar changes during parasite differentiation are mediated by an autophagy-like process. We have identified ATG8 in Plasmodium liver forms as an important effector that controls the development and fate of organelles, e.g., the clearance of micronemes that are required for hepatocyte invasion and the expansion of the apicoplast that produces many metabolites indispensable for parasite replication. Given the unconventional properties and the importance of ATG8 for parasite development in hepatocytes, targeting the parasite’s autophagic pathway may represent a novel approach to control malarial infections.
format article
author Christiane Voss
Karen Ehrenman
Godfree Mlambo
Satish Mishra
Kota Arun Kumar
John B. Sacci
Photini Sinnis
Isabelle Coppens
author_facet Christiane Voss
Karen Ehrenman
Godfree Mlambo
Satish Mishra
Kota Arun Kumar
John B. Sacci
Photini Sinnis
Isabelle Coppens
author_sort Christiane Voss
title Overexpression of <named-content content-type="genus-species">Plasmodium berghei</named-content> ATG8 by Liver Forms Leads to Cumulative Defects in Organelle Dynamics and to Generation of Noninfectious Merozoites
title_short Overexpression of <named-content content-type="genus-species">Plasmodium berghei</named-content> ATG8 by Liver Forms Leads to Cumulative Defects in Organelle Dynamics and to Generation of Noninfectious Merozoites
title_full Overexpression of <named-content content-type="genus-species">Plasmodium berghei</named-content> ATG8 by Liver Forms Leads to Cumulative Defects in Organelle Dynamics and to Generation of Noninfectious Merozoites
title_fullStr Overexpression of <named-content content-type="genus-species">Plasmodium berghei</named-content> ATG8 by Liver Forms Leads to Cumulative Defects in Organelle Dynamics and to Generation of Noninfectious Merozoites
title_full_unstemmed Overexpression of <named-content content-type="genus-species">Plasmodium berghei</named-content> ATG8 by Liver Forms Leads to Cumulative Defects in Organelle Dynamics and to Generation of Noninfectious Merozoites
title_sort overexpression of <named-content content-type="genus-species">plasmodium berghei</named-content> atg8 by liver forms leads to cumulative defects in organelle dynamics and to generation of noninfectious merozoites
publisher American Society for Microbiology
publishDate 2016
url https://doaj.org/article/9f9ba6d42b60419d9b7eb61fbe9c7b10
work_keys_str_mv AT christianevoss overexpressionofnamedcontentcontenttypegenusspeciesplasmodiumbergheinamedcontentatg8byliverformsleadstocumulativedefectsinorganelledynamicsandtogenerationofnoninfectiousmerozoites
AT karenehrenman overexpressionofnamedcontentcontenttypegenusspeciesplasmodiumbergheinamedcontentatg8byliverformsleadstocumulativedefectsinorganelledynamicsandtogenerationofnoninfectiousmerozoites
AT godfreemlambo overexpressionofnamedcontentcontenttypegenusspeciesplasmodiumbergheinamedcontentatg8byliverformsleadstocumulativedefectsinorganelledynamicsandtogenerationofnoninfectiousmerozoites
AT satishmishra overexpressionofnamedcontentcontenttypegenusspeciesplasmodiumbergheinamedcontentatg8byliverformsleadstocumulativedefectsinorganelledynamicsandtogenerationofnoninfectiousmerozoites
AT kotaarunkumar overexpressionofnamedcontentcontenttypegenusspeciesplasmodiumbergheinamedcontentatg8byliverformsleadstocumulativedefectsinorganelledynamicsandtogenerationofnoninfectiousmerozoites
AT johnbsacci overexpressionofnamedcontentcontenttypegenusspeciesplasmodiumbergheinamedcontentatg8byliverformsleadstocumulativedefectsinorganelledynamicsandtogenerationofnoninfectiousmerozoites
AT photinisinnis overexpressionofnamedcontentcontenttypegenusspeciesplasmodiumbergheinamedcontentatg8byliverformsleadstocumulativedefectsinorganelledynamicsandtogenerationofnoninfectiousmerozoites
AT isabellecoppens overexpressionofnamedcontentcontenttypegenusspeciesplasmodiumbergheinamedcontentatg8byliverformsleadstocumulativedefectsinorganelledynamicsandtogenerationofnoninfectiousmerozoites
_version_ 1718427475813335040