Stem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro.

Cystinosis is a rare disease caused by homozygous mutations of the CTNS gene, encoding a cystine efflux channel in the lysosomal membrane. In Ctns knockout mice, the pathologic intralysosomal accumulation of cystine that drives progressive organ damage can be reversed by infusion of wildtype bone ma...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Diana M Iglesias, Reyhan El-Kares, Anna Taranta, Francesco Bellomo, Francesco Emma, Martine Besouw, Elena Levtchenko, Jaan Toelen, Lambertus van den Heuvel, Leelee Chu, Jing Zhao, Yoon Kow Young, Nicoletta Eliopoulos, Paul Goodyer
Formato: article
Lenguaje:EN
Publicado: Public Library of Science (PLoS) 2012
Materias:
R
Q
Acceso en línea:https://doaj.org/article/353a18b5afee4d52adc1fd7c0dda280d
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:353a18b5afee4d52adc1fd7c0dda280d
record_format dspace
spelling oai:doaj.org-article:353a18b5afee4d52adc1fd7c0dda280d2021-11-18T07:08:57ZStem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro.1932-620310.1371/journal.pone.0042840https://doaj.org/article/353a18b5afee4d52adc1fd7c0dda280d2012-01-01T00:00:00Zhttps://www.ncbi.nlm.nih.gov/pmc/articles/pmid/22912749/pdf/?tool=EBIhttps://doaj.org/toc/1932-6203Cystinosis is a rare disease caused by homozygous mutations of the CTNS gene, encoding a cystine efflux channel in the lysosomal membrane. In Ctns knockout mice, the pathologic intralysosomal accumulation of cystine that drives progressive organ damage can be reversed by infusion of wildtype bone marrow-derived stem cells, but the mechanism involved is unclear since the exogeneous stem cells are rarely integrated into renal tubules. Here we show that human mesenchymal stem cells, from amniotic fluid or bone marrow, reduce pathologic cystine accumulation in co-cultured CTNS mutant fibroblasts or proximal tubular cells from cystinosis patients. This paracrine effect is associated with release into the culture medium of stem cell microvesicles (100-400 nm diameter) containing wildtype cystinosin protein and CTNS mRNA. Isolated stem cell microvesicles reduce target cell cystine accumulation in a dose-dependent, Annexin V-sensitive manner. Microvesicles from stem cells expressing CTNS(Red) transfer tagged CTNS protein to the lysosome/endosome compartment of cystinotic fibroblasts. Our observations suggest that exogenous stem cells may reprogram the biology of mutant tissues by direct microvesicle transfer of membrane-associated wildtype molecules.Diana M IglesiasReyhan El-KaresAnna TarantaFrancesco BellomoFrancesco EmmaMartine BesouwElena LevtchenkoJaan ToelenLambertus van den HeuvelLeelee ChuJing ZhaoYoon Kow YoungNicoletta EliopoulosPaul GoodyerPublic Library of Science (PLoS)articleMedicineRScienceQENPLoS ONE, Vol 7, Iss 8, p e42840 (2012)
institution DOAJ
collection DOAJ
language EN
topic Medicine
R
Science
Q
spellingShingle Medicine
R
Science
Q
Diana M Iglesias
Reyhan El-Kares
Anna Taranta
Francesco Bellomo
Francesco Emma
Martine Besouw
Elena Levtchenko
Jaan Toelen
Lambertus van den Heuvel
Leelee Chu
Jing Zhao
Yoon Kow Young
Nicoletta Eliopoulos
Paul Goodyer
Stem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro.
description Cystinosis is a rare disease caused by homozygous mutations of the CTNS gene, encoding a cystine efflux channel in the lysosomal membrane. In Ctns knockout mice, the pathologic intralysosomal accumulation of cystine that drives progressive organ damage can be reversed by infusion of wildtype bone marrow-derived stem cells, but the mechanism involved is unclear since the exogeneous stem cells are rarely integrated into renal tubules. Here we show that human mesenchymal stem cells, from amniotic fluid or bone marrow, reduce pathologic cystine accumulation in co-cultured CTNS mutant fibroblasts or proximal tubular cells from cystinosis patients. This paracrine effect is associated with release into the culture medium of stem cell microvesicles (100-400 nm diameter) containing wildtype cystinosin protein and CTNS mRNA. Isolated stem cell microvesicles reduce target cell cystine accumulation in a dose-dependent, Annexin V-sensitive manner. Microvesicles from stem cells expressing CTNS(Red) transfer tagged CTNS protein to the lysosome/endosome compartment of cystinotic fibroblasts. Our observations suggest that exogenous stem cells may reprogram the biology of mutant tissues by direct microvesicle transfer of membrane-associated wildtype molecules.
format article
author Diana M Iglesias
Reyhan El-Kares
Anna Taranta
Francesco Bellomo
Francesco Emma
Martine Besouw
Elena Levtchenko
Jaan Toelen
Lambertus van den Heuvel
Leelee Chu
Jing Zhao
Yoon Kow Young
Nicoletta Eliopoulos
Paul Goodyer
author_facet Diana M Iglesias
Reyhan El-Kares
Anna Taranta
Francesco Bellomo
Francesco Emma
Martine Besouw
Elena Levtchenko
Jaan Toelen
Lambertus van den Heuvel
Leelee Chu
Jing Zhao
Yoon Kow Young
Nicoletta Eliopoulos
Paul Goodyer
author_sort Diana M Iglesias
title Stem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro.
title_short Stem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro.
title_full Stem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro.
title_fullStr Stem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro.
title_full_unstemmed Stem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro.
title_sort stem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro.
publisher Public Library of Science (PLoS)
publishDate 2012
url https://doaj.org/article/353a18b5afee4d52adc1fd7c0dda280d
work_keys_str_mv AT dianamiglesias stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT reyhanelkares stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT annataranta stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT francescobellomo stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT francescoemma stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT martinebesouw stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT elenalevtchenko stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT jaantoelen stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT lambertusvandenheuvel stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT leeleechu stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT jingzhao stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT yoonkowyoung stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT nicolettaeliopoulos stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
AT paulgoodyer stemcellmicrovesiclestransfercystinosintohumancystinoticcellsandreducecystineaccumulationinvitro
_version_ 1718423844871471104