Aberrant Autophagy Impacts Growth and Multicellular Development in a Dictyostelium Knockout Model of CLN5 Disease

Mutations in CLN5 cause a subtype of neuronal ceroid lipofuscinosis (NCL) called CLN5 disease. While the precise role of CLN5 in NCL pathogenesis is not known, recent work revealed that the protein has glycoside hydrolase activity. Previous work on the Dictyostelium discoideum homolog of human CLN5,...

Full description

Bibliographic Details
Main Authors: Meagan D. McLaren, Sabateeshan Mathavarajah, William D. Kim, Shyong Q. Yap, Robert J. Huber
Format: Article
Language:English
Published: Frontiers Media S.A. 2021-07-01
Series:Frontiers in Cell and Developmental Biology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fcell.2021.657406/full
id doaj-b014e2beab594a958c545b9d4b975230
record_format Article
spelling doaj-b014e2beab594a958c545b9d4b9752302021-07-05T08:22:51ZengFrontiers Media S.A.Frontiers in Cell and Developmental Biology2296-634X2021-07-01910.3389/fcell.2021.657406657406Aberrant Autophagy Impacts Growth and Multicellular Development in a Dictyostelium Knockout Model of CLN5 DiseaseMeagan D. McLaren0Sabateeshan Mathavarajah1William D. Kim2Shyong Q. Yap3Robert J. Huber4Environmental and Life Sciences Graduate Program, Trent University, Peterborough, ON, CanadaDepartment of Biology, Trent University, Peterborough, ON, CanadaEnvironmental and Life Sciences Graduate Program, Trent University, Peterborough, ON, CanadaEnvironmental and Life Sciences Graduate Program, Trent University, Peterborough, ON, CanadaDepartment of Biology, Trent University, Peterborough, ON, CanadaMutations in CLN5 cause a subtype of neuronal ceroid lipofuscinosis (NCL) called CLN5 disease. While the precise role of CLN5 in NCL pathogenesis is not known, recent work revealed that the protein has glycoside hydrolase activity. Previous work on the Dictyostelium discoideum homolog of human CLN5, Cln5, revealed its secretion during the early stages of development and its role in regulating cell adhesion and cAMP-mediated chemotaxis. Here, we used Dictyostelium to examine the effect of cln5-deficiency on various growth and developmental processes during the life cycle. During growth, cln5– cells displayed reduced cell proliferation, cytokinesis, viability, and folic acid-mediated chemotaxis. In addition, the growth of cln5– cells was severely impaired in nutrient-limiting media. Based on these findings, we assessed autophagic flux in growth-phase cells and observed that loss of cln5 increased the number of autophagosomes suggesting that the basal level of autophagy was increased in cln5– cells. Similarly, loss of cln5 increased the amounts of ubiquitin-positive proteins. During the early stages of multicellular development, the aggregation of cln5– cells was delayed and loss of the autophagy genes, atg1 and atg9, reduced the extracellular amount of Cln5. We also observed an increased amount of intracellular Cln5 in cells lacking the Dictyostelium homolog of the human glycoside hydrolase, hexosaminidase A (HEXA), further supporting the glycoside hydrolase activity of Cln5. This observation was also supported by our finding that CLN5 and HEXA expression are highly correlated in human tissues. Following mound formation, cln5– development was precocious and loss of cln5 affected spore morphology, germination, and viability. When cln5– cells were developed in the presence of the autophagy inhibitor ammonium chloride, the formation of multicellular structures was impaired, and the size of cln5– slugs was reduced relative to WT slugs. These results, coupled with the aberrant autophagic flux observed in cln5– cells during growth, support a role for Cln5 in autophagy during the Dictyostelium life cycle. In total, this study highlights the multifaceted role of Cln5 in Dictyostelium and provides insight into the pathological mechanisms that may underlie CLN5 disease.https://www.frontiersin.org/articles/10.3389/fcell.2021.657406/fullautophagyBatten diseaseCLN5developmentDictyostelium discoideumglycoside hydrolase
collection DOAJ
language English
format Article
sources DOAJ
author Meagan D. McLaren
Sabateeshan Mathavarajah
William D. Kim
Shyong Q. Yap
Robert J. Huber
spellingShingle Meagan D. McLaren
Sabateeshan Mathavarajah
William D. Kim
Shyong Q. Yap
Robert J. Huber
Aberrant Autophagy Impacts Growth and Multicellular Development in a Dictyostelium Knockout Model of CLN5 Disease
Frontiers in Cell and Developmental Biology
autophagy
Batten disease
CLN5
development
Dictyostelium discoideum
glycoside hydrolase
author_facet Meagan D. McLaren
Sabateeshan Mathavarajah
William D. Kim
Shyong Q. Yap
Robert J. Huber
author_sort Meagan D. McLaren
title Aberrant Autophagy Impacts Growth and Multicellular Development in a Dictyostelium Knockout Model of CLN5 Disease
title_short Aberrant Autophagy Impacts Growth and Multicellular Development in a Dictyostelium Knockout Model of CLN5 Disease
title_full Aberrant Autophagy Impacts Growth and Multicellular Development in a Dictyostelium Knockout Model of CLN5 Disease
title_fullStr Aberrant Autophagy Impacts Growth and Multicellular Development in a Dictyostelium Knockout Model of CLN5 Disease
title_full_unstemmed Aberrant Autophagy Impacts Growth and Multicellular Development in a Dictyostelium Knockout Model of CLN5 Disease
title_sort aberrant autophagy impacts growth and multicellular development in a dictyostelium knockout model of cln5 disease
publisher Frontiers Media S.A.
series Frontiers in Cell and Developmental Biology
issn 2296-634X
publishDate 2021-07-01
description Mutations in CLN5 cause a subtype of neuronal ceroid lipofuscinosis (NCL) called CLN5 disease. While the precise role of CLN5 in NCL pathogenesis is not known, recent work revealed that the protein has glycoside hydrolase activity. Previous work on the Dictyostelium discoideum homolog of human CLN5, Cln5, revealed its secretion during the early stages of development and its role in regulating cell adhesion and cAMP-mediated chemotaxis. Here, we used Dictyostelium to examine the effect of cln5-deficiency on various growth and developmental processes during the life cycle. During growth, cln5– cells displayed reduced cell proliferation, cytokinesis, viability, and folic acid-mediated chemotaxis. In addition, the growth of cln5– cells was severely impaired in nutrient-limiting media. Based on these findings, we assessed autophagic flux in growth-phase cells and observed that loss of cln5 increased the number of autophagosomes suggesting that the basal level of autophagy was increased in cln5– cells. Similarly, loss of cln5 increased the amounts of ubiquitin-positive proteins. During the early stages of multicellular development, the aggregation of cln5– cells was delayed and loss of the autophagy genes, atg1 and atg9, reduced the extracellular amount of Cln5. We also observed an increased amount of intracellular Cln5 in cells lacking the Dictyostelium homolog of the human glycoside hydrolase, hexosaminidase A (HEXA), further supporting the glycoside hydrolase activity of Cln5. This observation was also supported by our finding that CLN5 and HEXA expression are highly correlated in human tissues. Following mound formation, cln5– development was precocious and loss of cln5 affected spore morphology, germination, and viability. When cln5– cells were developed in the presence of the autophagy inhibitor ammonium chloride, the formation of multicellular structures was impaired, and the size of cln5– slugs was reduced relative to WT slugs. These results, coupled with the aberrant autophagic flux observed in cln5– cells during growth, support a role for Cln5 in autophagy during the Dictyostelium life cycle. In total, this study highlights the multifaceted role of Cln5 in Dictyostelium and provides insight into the pathological mechanisms that may underlie CLN5 disease.
topic autophagy
Batten disease
CLN5
development
Dictyostelium discoideum
glycoside hydrolase
url https://www.frontiersin.org/articles/10.3389/fcell.2021.657406/full
work_keys_str_mv AT meagandmclaren aberrantautophagyimpactsgrowthandmulticellulardevelopmentinadictyosteliumknockoutmodelofcln5disease
AT sabateeshanmathavarajah aberrantautophagyimpactsgrowthandmulticellulardevelopmentinadictyosteliumknockoutmodelofcln5disease
AT williamdkim aberrantautophagyimpactsgrowthandmulticellulardevelopmentinadictyosteliumknockoutmodelofcln5disease
AT shyongqyap aberrantautophagyimpactsgrowthandmulticellulardevelopmentinadictyosteliumknockoutmodelofcln5disease
AT robertjhuber aberrantautophagyimpactsgrowthandmulticellulardevelopmentinadictyosteliumknockoutmodelofcln5disease
_version_ 1721318778243907584