Autophagy Promotes the Survival of Adipose Mesenchymal Stem/Stromal Cells and Enhances Their Therapeutic Effects in Cisplatin-Induced Liver Injury via Modulating TGF-β1/Smad and PI3K/AKT Signaling Pathways

Autophagy is a key metabolic process where cells can recycle its proteins and organelles to regenerate its own cellular building blocks. Chemotherapy is indispensable for cancer treatment but associated with various side-effects, including organ damage. Stem cell-based therapy is a promising approac...

Full description

Bibliographic Details
Main Authors: Eman Mohamad El Nashar, Mansour Abdullah Alghamdi, Wardah Abdullah Alasmari, Mohamed M. A. Hussein, Eman Hamza, Reham Ismail Taha, Mona M. Ahmed, Khulood Mohammed Al-Khater, Ahmed Abdelfattah-Hassan
Format: Article
Language:English
Published: MDPI AG 2021-09-01
Series:Cells
Subjects:
Online Access:https://www.mdpi.com/2073-4409/10/9/2475
_version_ 1797519814340640768
author Eman Mohamad El Nashar
Mansour Abdullah Alghamdi
Wardah Abdullah Alasmari
Mohamed M. A. Hussein
Eman Hamza
Reham Ismail Taha
Mona M. Ahmed
Khulood Mohammed Al-Khater
Ahmed Abdelfattah-Hassan
author_facet Eman Mohamad El Nashar
Mansour Abdullah Alghamdi
Wardah Abdullah Alasmari
Mohamed M. A. Hussein
Eman Hamza
Reham Ismail Taha
Mona M. Ahmed
Khulood Mohammed Al-Khater
Ahmed Abdelfattah-Hassan
author_sort Eman Mohamad El Nashar
collection DOAJ
description Autophagy is a key metabolic process where cells can recycle its proteins and organelles to regenerate its own cellular building blocks. Chemotherapy is indispensable for cancer treatment but associated with various side-effects, including organ damage. Stem cell-based therapy is a promising approach for reducing chemotherapeutic side effects, however, one of its main culprits is the poor survival of transplanted stem cells in damaged tissues. Here, we aimed to test the effects of activating autophagy in adipose-derived mesenchymal stem/stromal cells (ADSCs) on the survival of ADSCs, and their therapeutic value in cisplatin-induced liver injury model. Autophagy was activated in ADSCs by rapamycin (50 nM/L) for two hours before transplantation and were compared to non-preconditioned ADSCs. Rapamycin preconditioning resulted in activated autophagy and improved survival of ADSCs achieved by increased autophagosomes, upregulated autophagy-specific LC3-II gene, decreased protein degradation/ubiquitination by downregulated p62 gene, downregulated mTOR gene, and finally, upregulated antiapoptotic BCL-2 gene. In addition, autophagic ADSCs transplantation in the cisplatin liver injury model, liver biochemical parameters (AST, ALT and albumin), lipid peroxidation (MDA), antioxidant profile (SOD and GPX) and histopathological picture were improved, approaching near-normal conditions. These promising autophagic ADSCs effects were achieved by modulation of components in TGF-β1/Smad and PI3K-AKT signaling pathways, besides reducing NF-κB gene expression (marker for inflammation), reducing TGF-β1 levels (marker for fibrosis) and increasing SDF-1 levels (liver regeneration marker) in liver. Therefore, current results highlight the importance of autophagy in augmenting the therapeutic potential of stem cell therapy in alleviating cisplatin-associated liver damage and opens the path for improved cell-based therapies, in general, and with chemotherapeutics, in particular.
first_indexed 2024-03-10T07:48:07Z
format Article
id doaj.art-285d3410d018437f94e1f9b7a607f07c
institution Directory Open Access Journal
issn 2073-4409
language English
last_indexed 2024-03-10T07:48:07Z
publishDate 2021-09-01
publisher MDPI AG
record_format Article
series Cells
spelling doaj.art-285d3410d018437f94e1f9b7a607f07c2023-11-22T12:26:49ZengMDPI AGCells2073-44092021-09-01109247510.3390/cells10092475Autophagy Promotes the Survival of Adipose Mesenchymal Stem/Stromal Cells and Enhances Their Therapeutic Effects in Cisplatin-Induced Liver Injury via Modulating TGF-β1/Smad and PI3K/AKT Signaling PathwaysEman Mohamad El Nashar0Mansour Abdullah Alghamdi1Wardah Abdullah Alasmari2Mohamed M. A. Hussein3Eman Hamza4Reham Ismail Taha5Mona M. Ahmed6Khulood Mohammed Al-Khater7Ahmed Abdelfattah-Hassan8Department of Anatomy, College of Medicine, King Khalid University, Abha 61421, Saudi ArabiaDepartment of Anatomy, College of Medicine, King Khalid University, Abha 61421, Saudi ArabiaDepartment of Anatomy, Faculty of Medicine, Umm Al-Qura University, Makkah 24230, Saudi ArabiaBiochemistry Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, EgyptDepartment of Medical Biochemistry, Faculty of Medicine, Mansoura University, Mansoura 35516, EgyptDepartment of Anatomy, Faculty of Medicine, Mansoura University, Mansoura 35516, EgyptForensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, EgyptDepartment of Anatomy, College of Medicine, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi ArabiaDepartment of Anatomy and Embryology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, EgyptAutophagy is a key metabolic process where cells can recycle its proteins and organelles to regenerate its own cellular building blocks. Chemotherapy is indispensable for cancer treatment but associated with various side-effects, including organ damage. Stem cell-based therapy is a promising approach for reducing chemotherapeutic side effects, however, one of its main culprits is the poor survival of transplanted stem cells in damaged tissues. Here, we aimed to test the effects of activating autophagy in adipose-derived mesenchymal stem/stromal cells (ADSCs) on the survival of ADSCs, and their therapeutic value in cisplatin-induced liver injury model. Autophagy was activated in ADSCs by rapamycin (50 nM/L) for two hours before transplantation and were compared to non-preconditioned ADSCs. Rapamycin preconditioning resulted in activated autophagy and improved survival of ADSCs achieved by increased autophagosomes, upregulated autophagy-specific LC3-II gene, decreased protein degradation/ubiquitination by downregulated p62 gene, downregulated mTOR gene, and finally, upregulated antiapoptotic BCL-2 gene. In addition, autophagic ADSCs transplantation in the cisplatin liver injury model, liver biochemical parameters (AST, ALT and albumin), lipid peroxidation (MDA), antioxidant profile (SOD and GPX) and histopathological picture were improved, approaching near-normal conditions. These promising autophagic ADSCs effects were achieved by modulation of components in TGF-β1/Smad and PI3K-AKT signaling pathways, besides reducing NF-κB gene expression (marker for inflammation), reducing TGF-β1 levels (marker for fibrosis) and increasing SDF-1 levels (liver regeneration marker) in liver. Therefore, current results highlight the importance of autophagy in augmenting the therapeutic potential of stem cell therapy in alleviating cisplatin-associated liver damage and opens the path for improved cell-based therapies, in general, and with chemotherapeutics, in particular.https://www.mdpi.com/2073-4409/10/9/2475ADSCscell-based therapyrapamycinapoptosisliver damagehepatoprotective pathways
spellingShingle Eman Mohamad El Nashar
Mansour Abdullah Alghamdi
Wardah Abdullah Alasmari
Mohamed M. A. Hussein
Eman Hamza
Reham Ismail Taha
Mona M. Ahmed
Khulood Mohammed Al-Khater
Ahmed Abdelfattah-Hassan
Autophagy Promotes the Survival of Adipose Mesenchymal Stem/Stromal Cells and Enhances Their Therapeutic Effects in Cisplatin-Induced Liver Injury via Modulating TGF-β1/Smad and PI3K/AKT Signaling Pathways
Cells
ADSCs
cell-based therapy
rapamycin
apoptosis
liver damage
hepatoprotective pathways
title Autophagy Promotes the Survival of Adipose Mesenchymal Stem/Stromal Cells and Enhances Their Therapeutic Effects in Cisplatin-Induced Liver Injury via Modulating TGF-β1/Smad and PI3K/AKT Signaling Pathways
title_full Autophagy Promotes the Survival of Adipose Mesenchymal Stem/Stromal Cells and Enhances Their Therapeutic Effects in Cisplatin-Induced Liver Injury via Modulating TGF-β1/Smad and PI3K/AKT Signaling Pathways
title_fullStr Autophagy Promotes the Survival of Adipose Mesenchymal Stem/Stromal Cells and Enhances Their Therapeutic Effects in Cisplatin-Induced Liver Injury via Modulating TGF-β1/Smad and PI3K/AKT Signaling Pathways
title_full_unstemmed Autophagy Promotes the Survival of Adipose Mesenchymal Stem/Stromal Cells and Enhances Their Therapeutic Effects in Cisplatin-Induced Liver Injury via Modulating TGF-β1/Smad and PI3K/AKT Signaling Pathways
title_short Autophagy Promotes the Survival of Adipose Mesenchymal Stem/Stromal Cells and Enhances Their Therapeutic Effects in Cisplatin-Induced Liver Injury via Modulating TGF-β1/Smad and PI3K/AKT Signaling Pathways
title_sort autophagy promotes the survival of adipose mesenchymal stem stromal cells and enhances their therapeutic effects in cisplatin induced liver injury via modulating tgf β1 smad and pi3k akt signaling pathways
topic ADSCs
cell-based therapy
rapamycin
apoptosis
liver damage
hepatoprotective pathways
url https://www.mdpi.com/2073-4409/10/9/2475
work_keys_str_mv AT emanmohamadelnashar autophagypromotesthesurvivalofadiposemesenchymalstemstromalcellsandenhancestheirtherapeuticeffectsincisplatininducedliverinjuryviamodulatingtgfb1smadandpi3kaktsignalingpathways
AT mansourabdullahalghamdi autophagypromotesthesurvivalofadiposemesenchymalstemstromalcellsandenhancestheirtherapeuticeffectsincisplatininducedliverinjuryviamodulatingtgfb1smadandpi3kaktsignalingpathways
AT wardahabdullahalasmari autophagypromotesthesurvivalofadiposemesenchymalstemstromalcellsandenhancestheirtherapeuticeffectsincisplatininducedliverinjuryviamodulatingtgfb1smadandpi3kaktsignalingpathways
AT mohamedmahussein autophagypromotesthesurvivalofadiposemesenchymalstemstromalcellsandenhancestheirtherapeuticeffectsincisplatininducedliverinjuryviamodulatingtgfb1smadandpi3kaktsignalingpathways
AT emanhamza autophagypromotesthesurvivalofadiposemesenchymalstemstromalcellsandenhancestheirtherapeuticeffectsincisplatininducedliverinjuryviamodulatingtgfb1smadandpi3kaktsignalingpathways
AT rehamismailtaha autophagypromotesthesurvivalofadiposemesenchymalstemstromalcellsandenhancestheirtherapeuticeffectsincisplatininducedliverinjuryviamodulatingtgfb1smadandpi3kaktsignalingpathways
AT monamahmed autophagypromotesthesurvivalofadiposemesenchymalstemstromalcellsandenhancestheirtherapeuticeffectsincisplatininducedliverinjuryviamodulatingtgfb1smadandpi3kaktsignalingpathways
AT khuloodmohammedalkhater autophagypromotesthesurvivalofadiposemesenchymalstemstromalcellsandenhancestheirtherapeuticeffectsincisplatininducedliverinjuryviamodulatingtgfb1smadandpi3kaktsignalingpathways
AT ahmedabdelfattahhassan autophagypromotesthesurvivalofadiposemesenchymalstemstromalcellsandenhancestheirtherapeuticeffectsincisplatininducedliverinjuryviamodulatingtgfb1smadandpi3kaktsignalingpathways