A-Kinase Anchor Protein 1 deficiency causes mitochondrial dysfunction in mouse model of hyperoxia induced acute lung injury

Background: Critically ill patients on supplemental oxygen therapy eventually develop acute lung injury (ALI). Reactive oxygen species (ROS) produced during ALI perturbs the mitochondrial dynamics resulting in cellular damage. Genetic deletion of the mitochondrial A-kinase anchoring protein 1 (Akap1...

Full description

Bibliographic Details
Main Authors: Ramani Soundararajan, Helena Hernández-Cuervo, Timothy M Stearns, Anthony J Griswold, Sahebgowda Sidramagowda Patil, Jutaro Fukumoto, Venkata Ramireddy Narala, Lakshmi Galam, Richard Lockey, Narasaiah Kolliputi
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-10-01
Series:Frontiers in Pharmacology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fphar.2022.980723/full
_version_ 1811181834116005888
author Ramani Soundararajan
Helena Hernández-Cuervo
Helena Hernández-Cuervo
Timothy M Stearns
Anthony J Griswold
Sahebgowda Sidramagowda Patil
Jutaro Fukumoto
Venkata Ramireddy Narala
Lakshmi Galam
Richard Lockey
Narasaiah Kolliputi
Narasaiah Kolliputi
author_facet Ramani Soundararajan
Helena Hernández-Cuervo
Helena Hernández-Cuervo
Timothy M Stearns
Anthony J Griswold
Sahebgowda Sidramagowda Patil
Jutaro Fukumoto
Venkata Ramireddy Narala
Lakshmi Galam
Richard Lockey
Narasaiah Kolliputi
Narasaiah Kolliputi
author_sort Ramani Soundararajan
collection DOAJ
description Background: Critically ill patients on supplemental oxygen therapy eventually develop acute lung injury (ALI). Reactive oxygen species (ROS) produced during ALI perturbs the mitochondrial dynamics resulting in cellular damage. Genetic deletion of the mitochondrial A-kinase anchoring protein 1 (Akap1) in mice resulted in mitochondrial damage, Endoplasmic reticulum (ER) stress, increased expression of mitophagy proteins and pro-inflammatory cytokines, exacerbating hyperoxia-induced Acute Lung Injury (HALI).Objective: Despite a strong causal link between mitochondrial dysfunction and HALI, the mechanisms governing the disease progression at the transcriptome level is unknown.Methods: In this study, RNA sequencing (RNA-seq) analysis was carried out using the lungs of Akap1 knockout (Akap1−/−) mice exposed to normoxia or 48 h of hyperoxia followed by quantitative real time PCR and Ingenuity pathway analysis (IPA). Western blot analysis assessed mitochondrial dysfunction, OXPHOS complex (I-V), apoptosis and antioxidant proteins. Mitochondrial enzymatic assays was used to measure the aconitase, fumarase, citrate synthase activities in isolated mitochondria from Akap1−/− vs. Wt mice exposed to hyperoxia.Results: Transcriptome analysis of Akap1−/− exposed to hyperoxia reveals increases in transcripts encoding electron transport chain (ETC) and tricarboxylic acid cycle (TCA) proteins. Ingenuity pathway analysis (IPA) shows enrichment of mitochondrial dysfunction and oxidative phosphorylation in Akap1−/− mice. Loss of AKAP1, coupled with oxidant injury, significantly decreases the activities of TCA enzymes. Mechanistically, a significant loss of dynamin-related protein 1 (Drp1) phosphorylation at the protein kinase A (PKA) site Serine 637 (Ser637), decreases in Akt phosphorylation at Serine 437 (Ser47) and increase in the expression of pro-apoptotic protein Bax indicate mitochondrial dysfunction. Heme oxygenase-1 (HO-1) levels significantly increased in CD68 positive alveolar macrophages in Akap1−/− lungs, suggesting a strong antioxidant response to hyperoxia.Conclusion: Overall these results suggest that AKAP1 overexpression and modulation of Drp1 phosphorylation at Ser637 is an important therapeutic strategy for acute lung injury.
first_indexed 2024-04-11T09:23:34Z
format Article
id doaj.art-60d6092c74c549c8962d337265289e32
institution Directory Open Access Journal
issn 1663-9812
language English
last_indexed 2024-04-11T09:23:34Z
publishDate 2022-10-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Pharmacology
spelling doaj.art-60d6092c74c549c8962d337265289e322022-12-22T04:32:06ZengFrontiers Media S.A.Frontiers in Pharmacology1663-98122022-10-011310.3389/fphar.2022.980723980723A-Kinase Anchor Protein 1 deficiency causes mitochondrial dysfunction in mouse model of hyperoxia induced acute lung injuryRamani Soundararajan0Helena Hernández-Cuervo1Helena Hernández-Cuervo2Timothy M Stearns3Anthony J Griswold4Sahebgowda Sidramagowda Patil5Jutaro Fukumoto6Venkata Ramireddy Narala7Lakshmi Galam8Richard Lockey9Narasaiah Kolliputi10Narasaiah Kolliputi11Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United StatesDivision of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United StatesUniversity of South Florida, Department of Molecular Medicine, Morsani College of Medicine, Tampa, FL, United StatesJackson Laboratory, Bar Harbor, ME, United StatesJohn P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, United StatesDivision of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United StatesDivision of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United StatesDepartment of Zoology, Yogi Vemana University, Kadapa, AP, IndiaDivision of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United StatesDivision of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United StatesDivision of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United StatesUniversity of South Florida, Department of Molecular Medicine, Morsani College of Medicine, Tampa, FL, United StatesBackground: Critically ill patients on supplemental oxygen therapy eventually develop acute lung injury (ALI). Reactive oxygen species (ROS) produced during ALI perturbs the mitochondrial dynamics resulting in cellular damage. Genetic deletion of the mitochondrial A-kinase anchoring protein 1 (Akap1) in mice resulted in mitochondrial damage, Endoplasmic reticulum (ER) stress, increased expression of mitophagy proteins and pro-inflammatory cytokines, exacerbating hyperoxia-induced Acute Lung Injury (HALI).Objective: Despite a strong causal link between mitochondrial dysfunction and HALI, the mechanisms governing the disease progression at the transcriptome level is unknown.Methods: In this study, RNA sequencing (RNA-seq) analysis was carried out using the lungs of Akap1 knockout (Akap1−/−) mice exposed to normoxia or 48 h of hyperoxia followed by quantitative real time PCR and Ingenuity pathway analysis (IPA). Western blot analysis assessed mitochondrial dysfunction, OXPHOS complex (I-V), apoptosis and antioxidant proteins. Mitochondrial enzymatic assays was used to measure the aconitase, fumarase, citrate synthase activities in isolated mitochondria from Akap1−/− vs. Wt mice exposed to hyperoxia.Results: Transcriptome analysis of Akap1−/− exposed to hyperoxia reveals increases in transcripts encoding electron transport chain (ETC) and tricarboxylic acid cycle (TCA) proteins. Ingenuity pathway analysis (IPA) shows enrichment of mitochondrial dysfunction and oxidative phosphorylation in Akap1−/− mice. Loss of AKAP1, coupled with oxidant injury, significantly decreases the activities of TCA enzymes. Mechanistically, a significant loss of dynamin-related protein 1 (Drp1) phosphorylation at the protein kinase A (PKA) site Serine 637 (Ser637), decreases in Akt phosphorylation at Serine 437 (Ser47) and increase in the expression of pro-apoptotic protein Bax indicate mitochondrial dysfunction. Heme oxygenase-1 (HO-1) levels significantly increased in CD68 positive alveolar macrophages in Akap1−/− lungs, suggesting a strong antioxidant response to hyperoxia.Conclusion: Overall these results suggest that AKAP1 overexpression and modulation of Drp1 phosphorylation at Ser637 is an important therapeutic strategy for acute lung injury.https://www.frontiersin.org/articles/10.3389/fphar.2022.980723/fullAkap1hyperoxiamitochondrial dysfunctionAktDrp1ETS
spellingShingle Ramani Soundararajan
Helena Hernández-Cuervo
Helena Hernández-Cuervo
Timothy M Stearns
Anthony J Griswold
Sahebgowda Sidramagowda Patil
Jutaro Fukumoto
Venkata Ramireddy Narala
Lakshmi Galam
Richard Lockey
Narasaiah Kolliputi
Narasaiah Kolliputi
A-Kinase Anchor Protein 1 deficiency causes mitochondrial dysfunction in mouse model of hyperoxia induced acute lung injury
Frontiers in Pharmacology
Akap1
hyperoxia
mitochondrial dysfunction
Akt
Drp1
ETS
title A-Kinase Anchor Protein 1 deficiency causes mitochondrial dysfunction in mouse model of hyperoxia induced acute lung injury
title_full A-Kinase Anchor Protein 1 deficiency causes mitochondrial dysfunction in mouse model of hyperoxia induced acute lung injury
title_fullStr A-Kinase Anchor Protein 1 deficiency causes mitochondrial dysfunction in mouse model of hyperoxia induced acute lung injury
title_full_unstemmed A-Kinase Anchor Protein 1 deficiency causes mitochondrial dysfunction in mouse model of hyperoxia induced acute lung injury
title_short A-Kinase Anchor Protein 1 deficiency causes mitochondrial dysfunction in mouse model of hyperoxia induced acute lung injury
title_sort a kinase anchor protein 1 deficiency causes mitochondrial dysfunction in mouse model of hyperoxia induced acute lung injury
topic Akap1
hyperoxia
mitochondrial dysfunction
Akt
Drp1
ETS
url https://www.frontiersin.org/articles/10.3389/fphar.2022.980723/full
work_keys_str_mv AT ramanisoundararajan akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury
AT helenahernandezcuervo akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury
AT helenahernandezcuervo akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury
AT timothymstearns akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury
AT anthonyjgriswold akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury
AT sahebgowdasidramagowdapatil akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury
AT jutarofukumoto akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury
AT venkataramireddynarala akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury
AT lakshmigalam akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury
AT richardlockey akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury
AT narasaiahkolliputi akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury
AT narasaiahkolliputi akinaseanchorprotein1deficiencycausesmitochondrialdysfunctioninmousemodelofhyperoxiainducedacutelunginjury