Establishing a link between endothelial cell metabolism and vascular behaviour in a type 1 diabetes mouse model

Detalhes bibliográficos
Autor(a) principal: Silva, C
Data de Publicação: 2019
Outros Autores: Sampaio-Pinto, V, Andrade, S, Rodrigues, I, Costa, R, Guerreiro, SG, Carvalho, E, Pinto-do-Ó, P, Nascimento, DS, Soares, R
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
Texto Completo: https://hdl.handle.net/10216/136304
Resumo: Background/Aims: Vascular complications contribute significantly to the extensive morbidity and mortality rates observed in people with diabetes. Despite well known that the diabetic kidney and heart exhibit imbalanced angiogenesis, the mechanisms implicated in this angiogenic paradox remain unknown. In this study, we examined the angiogenic and metabolic gene expression profile (GEP) of endothelial cells (ECs) isolated from a mouse model with type1 diabetes mellitus (T1DM). Methods: ECs were isolated from kidneys and hearts of healthy and streptozocin (STZ)-treated mice. RNA was then extracted for molecular studies. GEP of 84 angiogenic and 84 AMP-activated Protein Kinase (AMPK)-dependent genes were examined by microarrays. Real time PCR confirmed the changes observed in significantly altered genes. Microvessel density (MVD) was analysed by immunohistochemistry, fibrosis was assessed by the Sirius red histological staining and connective tissue growth factor (CTGF) was quantified by ELISA. Results: The relative percentage of ECs and MVD were increased in the kidneys of T1DM animals whereas the opposite trend was observed in the hearts of diabetic mice. Accordingly, the majority of AMPK-associated genes were upregulated in kidneys and downregulated in hearts of these animals. Angiogenic GEP revealed significant differences in Tgfß, Notch signaling and Timp2 in both diabetic organs. These findings were in agreement with the angiogenesis histological assays. Fibrosis was augmented in both organs in diabetic as compared to healthy animals. Conclusion: Altogether, our findings indicate, for the first time, that T1DM heart and kidney ECs present opposite metabolic cues, which are accompanied by distinct angiogenic patterns. These findings enable the development of innovative organ-specific therapeutic strategies targeting diabetic-associated vascular disorders.
id RCAP_2e4aceb27171f05d78740604090e044e
oai_identifier_str oai:repositorio-aberto.up.pt:10216/136304
network_acronym_str RCAP
network_name_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository_id_str 7160
spelling Establishing a link between endothelial cell metabolism and vascular behaviour in a type 1 diabetes mouse modelCarbohydrate and lipid metabolismCell sortingEndothelium metabolismGenomicsMacrovascular complicationsMicroBackground/Aims: Vascular complications contribute significantly to the extensive morbidity and mortality rates observed in people with diabetes. Despite well known that the diabetic kidney and heart exhibit imbalanced angiogenesis, the mechanisms implicated in this angiogenic paradox remain unknown. In this study, we examined the angiogenic and metabolic gene expression profile (GEP) of endothelial cells (ECs) isolated from a mouse model with type1 diabetes mellitus (T1DM). Methods: ECs were isolated from kidneys and hearts of healthy and streptozocin (STZ)-treated mice. RNA was then extracted for molecular studies. GEP of 84 angiogenic and 84 AMP-activated Protein Kinase (AMPK)-dependent genes were examined by microarrays. Real time PCR confirmed the changes observed in significantly altered genes. Microvessel density (MVD) was analysed by immunohistochemistry, fibrosis was assessed by the Sirius red histological staining and connective tissue growth factor (CTGF) was quantified by ELISA. Results: The relative percentage of ECs and MVD were increased in the kidneys of T1DM animals whereas the opposite trend was observed in the hearts of diabetic mice. Accordingly, the majority of AMPK-associated genes were upregulated in kidneys and downregulated in hearts of these animals. Angiogenic GEP revealed significant differences in Tgfß, Notch signaling and Timp2 in both diabetic organs. These findings were in agreement with the angiogenesis histological assays. Fibrosis was augmented in both organs in diabetic as compared to healthy animals. Conclusion: Altogether, our findings indicate, for the first time, that T1DM heart and kidney ECs present opposite metabolic cues, which are accompanied by distinct angiogenic patterns. These findings enable the development of innovative organ-specific therapeutic strategies targeting diabetic-associated vascular disorders.Cell Physiol Biochem Press GmbH & Co KG20192019-01-01T00:00:00Zinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/articleapplication/pdfhttps://hdl.handle.net/10216/136304eng1015-898710.33594/000000036Silva, CSampaio-Pinto, VAndrade, SRodrigues, ICosta, RGuerreiro, SGCarvalho, EPinto-do-Ó, PNascimento, DSSoares, Rinfo:eu-repo/semantics/openAccessreponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãoinstacron:RCAAP2023-11-29T16:02:23Zoai:repositorio-aberto.up.pt:10216/136304Portal AgregadorONGhttps://www.rcaap.pt/oai/openaireopendoar:71602024-03-20T00:37:05.851643Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãofalse
dc.title.none.fl_str_mv Establishing a link between endothelial cell metabolism and vascular behaviour in a type 1 diabetes mouse model
title Establishing a link between endothelial cell metabolism and vascular behaviour in a type 1 diabetes mouse model
spellingShingle Establishing a link between endothelial cell metabolism and vascular behaviour in a type 1 diabetes mouse model
Silva, C
Carbohydrate and lipid metabolism
Cell sorting
Endothelium metabolism
Genomics
Macrovascular complications
Micro
title_short Establishing a link between endothelial cell metabolism and vascular behaviour in a type 1 diabetes mouse model
title_full Establishing a link between endothelial cell metabolism and vascular behaviour in a type 1 diabetes mouse model
title_fullStr Establishing a link between endothelial cell metabolism and vascular behaviour in a type 1 diabetes mouse model
title_full_unstemmed Establishing a link between endothelial cell metabolism and vascular behaviour in a type 1 diabetes mouse model
title_sort Establishing a link between endothelial cell metabolism and vascular behaviour in a type 1 diabetes mouse model
author Silva, C
author_facet Silva, C
Sampaio-Pinto, V
Andrade, S
Rodrigues, I
Costa, R
Guerreiro, SG
Carvalho, E
Pinto-do-Ó, P
Nascimento, DS
Soares, R
author_role author
author2 Sampaio-Pinto, V
Andrade, S
Rodrigues, I
Costa, R
Guerreiro, SG
Carvalho, E
Pinto-do-Ó, P
Nascimento, DS
Soares, R
author2_role author
author
author
author
author
author
author
author
author
dc.contributor.author.fl_str_mv Silva, C
Sampaio-Pinto, V
Andrade, S
Rodrigues, I
Costa, R
Guerreiro, SG
Carvalho, E
Pinto-do-Ó, P
Nascimento, DS
Soares, R
dc.subject.por.fl_str_mv Carbohydrate and lipid metabolism
Cell sorting
Endothelium metabolism
Genomics
Macrovascular complications
Micro
topic Carbohydrate and lipid metabolism
Cell sorting
Endothelium metabolism
Genomics
Macrovascular complications
Micro
description Background/Aims: Vascular complications contribute significantly to the extensive morbidity and mortality rates observed in people with diabetes. Despite well known that the diabetic kidney and heart exhibit imbalanced angiogenesis, the mechanisms implicated in this angiogenic paradox remain unknown. In this study, we examined the angiogenic and metabolic gene expression profile (GEP) of endothelial cells (ECs) isolated from a mouse model with type1 diabetes mellitus (T1DM). Methods: ECs were isolated from kidneys and hearts of healthy and streptozocin (STZ)-treated mice. RNA was then extracted for molecular studies. GEP of 84 angiogenic and 84 AMP-activated Protein Kinase (AMPK)-dependent genes were examined by microarrays. Real time PCR confirmed the changes observed in significantly altered genes. Microvessel density (MVD) was analysed by immunohistochemistry, fibrosis was assessed by the Sirius red histological staining and connective tissue growth factor (CTGF) was quantified by ELISA. Results: The relative percentage of ECs and MVD were increased in the kidneys of T1DM animals whereas the opposite trend was observed in the hearts of diabetic mice. Accordingly, the majority of AMPK-associated genes were upregulated in kidneys and downregulated in hearts of these animals. Angiogenic GEP revealed significant differences in Tgfß, Notch signaling and Timp2 in both diabetic organs. These findings were in agreement with the angiogenesis histological assays. Fibrosis was augmented in both organs in diabetic as compared to healthy animals. Conclusion: Altogether, our findings indicate, for the first time, that T1DM heart and kidney ECs present opposite metabolic cues, which are accompanied by distinct angiogenic patterns. These findings enable the development of innovative organ-specific therapeutic strategies targeting diabetic-associated vascular disorders.
publishDate 2019
dc.date.none.fl_str_mv 2019
2019-01-01T00:00:00Z
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/article
format article
status_str publishedVersion
dc.identifier.uri.fl_str_mv https://hdl.handle.net/10216/136304
url https://hdl.handle.net/10216/136304
dc.language.iso.fl_str_mv eng
language eng
dc.relation.none.fl_str_mv 1015-8987
10.33594/000000036
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.publisher.none.fl_str_mv Cell Physiol Biochem Press GmbH & Co KG
publisher.none.fl_str_mv Cell Physiol Biochem Press GmbH & Co KG
dc.source.none.fl_str_mv reponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron:RCAAP
instname_str Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron_str RCAAP
institution RCAAP
reponame_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
collection Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository.name.fl_str_mv Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
repository.mail.fl_str_mv
_version_ 1799136279089119232