1
|
Csergeová L, Krbušek D, Janoštiak R. CIP/KIP and INK4 families as hostages of oncogenic signaling. Cell Div 2024; 19:11. [PMID: 38561743 PMCID: PMC10985988 DOI: 10.1186/s13008-024-00115-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
CIP/KIP and INK4 families of Cyclin-dependent kinase inhibitors (CKIs) are well-established cell cycle regulatory proteins whose canonical function is binding to Cyclin-CDK complexes and altering their function. Initial experiments showed that these proteins negatively regulate cell cycle progression and thus are tumor suppressors in the context of molecular oncology. However, expanded research into the functions of these proteins showed that most of them have non-canonical functions, both cell cycle-dependent and independent, and can even act as tumor enhancers depending on their posttranslational modifications, subcellular localization, and cell state context. This review aims to provide an overview of canonical as well as non-canonical functions of CIP/KIP and INK4 families of CKIs, discuss the potential avenues to promote their tumor suppressor functions instead of tumor enhancing ones, and how they could be utilized to design improved treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Lucia Csergeová
- BIOCEV-First Faculty of Medicine, Charles University, Prague, Czechia
| | - David Krbušek
- BIOCEV-First Faculty of Medicine, Charles University, Prague, Czechia
| | | |
Collapse
|
2
|
Deng RM, Zhou J. The role of PI3K/AKT signaling pathway in myocardial ischemia-reperfusion injury. Int Immunopharmacol 2023; 123:110714. [PMID: 37523969 DOI: 10.1016/j.intimp.2023.110714] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 07/09/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Myocardial ischemia has a high incidence and mortality rate, and reperfusion is currently the standard intervention. However, reperfusion may lead to further myocardial damage, known as myocardial ischemia/reperfusion injury (MIRI). There are currently no effective clinical treatments for MIRI. The PI3K/Akt signaling pathway is involved in cardiovascular health and disease and plays an important role in reducing myocardial infarct size and restoring cardiac function after MIRI. Activation of the PI3K/Akt pathway provides myocardial protection through synergistic upregulation of antioxidant, anti-inflammatory, and autophagy activities and inhibition of mitochondrial dysfunction and cardiomyocyte apoptosis. Many studies have shown that PI3K/Akt has a significant protective effect against MIRI. Here, we reviewed the molecular regulation of PI3K/Akt in MIRI and summarized the molecular mechanism by which PI3K/Akt affects MIRI, the effects of ischemic preconditioning and ischemic postconditioning, and the role of related drugs or activators targeting PI3K/Akt in MIRI, providing novel insights for the formulation of myocardial protection strategies. This review provides evidence of the role of PI3K/Akt activation in MIRI and supports its use as a therapeutic target.
Collapse
Affiliation(s)
- Rui-Ming Deng
- Department of Anesthesiology, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; The Affiliated Ganzhou Hospital of Nanchang University, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China
| | - Juan Zhou
- Department of thyroid and Breast Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; The Affiliated Ganzhou Hospital of Nanchang University, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| |
Collapse
|
3
|
Dhaibar HA, Kamberov L, Carroll NG, Amatya S, Cosic D, Gomez-Torres O, Vital S, Sivandzade F, Bhalerao A, Mancuso S, Shen X, Nam H, Orr AW, Dudenbostel T, Bailey SR, Kevil CG, Cucullo L, Cruz-Topete D. Exposure to Stress Alters Cardiac Gene Expression and Exacerbates Myocardial Ischemic Injury in the Female Murine Heart. Int J Mol Sci 2023; 24:10994. [PMID: 37446174 PMCID: PMC10341935 DOI: 10.3390/ijms241310994] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Mental stress is a risk factor for myocardial infarction in women. The central hypothesis of this study is that restraint stress induces sex-specific changes in gene expression in the heart, which leads to an intensified response to ischemia/reperfusion injury due to the development of a pro-oxidative environment in female hearts. We challenged male and female C57BL/6 mice in a restraint stress model to mimic the effects of mental stress. Exposure to restraint stress led to sex differences in the expression of genes involved in cardiac hypertrophy, inflammation, and iron-dependent cell death (ferroptosis). Among those genes, we identified tumor protein p53 and cyclin-dependent kinase inhibitor 1A (p21), which have established controversial roles in ferroptosis. The exacerbated response to I/R injury in restraint-stressed females correlated with downregulation of p53 and nuclear factor erythroid 2-related factor 2 (Nrf2, a master regulator of the antioxidant response system-ARE). S-female hearts also showed increased superoxide levels, lipid peroxidation, and prostaglandin-endoperoxide synthase 2 (Ptgs2) expression (a hallmark of ferroptosis) compared with those of their male counterparts. Our study is the first to test the sex-specific impact of restraint stress on the heart in the setting of I/R and its outcome.
Collapse
Affiliation(s)
- Hemangini A. Dhaibar
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; (H.A.D.); (L.K.); (N.G.C.); (S.A.); (D.C.); (O.G.-T.); (S.V.)
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
| | - Lilly Kamberov
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; (H.A.D.); (L.K.); (N.G.C.); (S.A.); (D.C.); (O.G.-T.); (S.V.)
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
| | - Natalie G. Carroll
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; (H.A.D.); (L.K.); (N.G.C.); (S.A.); (D.C.); (O.G.-T.); (S.V.)
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
| | - Shripa Amatya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; (H.A.D.); (L.K.); (N.G.C.); (S.A.); (D.C.); (O.G.-T.); (S.V.)
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
| | - Dario Cosic
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; (H.A.D.); (L.K.); (N.G.C.); (S.A.); (D.C.); (O.G.-T.); (S.V.)
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
| | - Oscar Gomez-Torres
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; (H.A.D.); (L.K.); (N.G.C.); (S.A.); (D.C.); (O.G.-T.); (S.V.)
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla-La Mancha, Toledo 45004, Spain
| | - Shantel Vital
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; (H.A.D.); (L.K.); (N.G.C.); (S.A.); (D.C.); (O.G.-T.); (S.V.)
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
| | - Farzane Sivandzade
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (F.S.); (A.B.); (S.M.)
- Department of Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Aditya Bhalerao
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (F.S.); (A.B.); (S.M.)
- Department of Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Salvatore Mancuso
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (F.S.); (A.B.); (S.M.)
- Department of Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Xinggui Shen
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Hyung Nam
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
- Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - A. Wayne Orr
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Tanja Dudenbostel
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
- LSU Health Sciences Center, Department of Internal Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Steven R. Bailey
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
- LSU Health Sciences Center, Department of Internal Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Christopher G. Kevil
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA
| | - Luca Cucullo
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA; (F.S.); (A.B.); (S.M.)
| | - Diana Cruz-Topete
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA 71103, USA; (H.A.D.); (L.K.); (N.G.C.); (S.A.); (D.C.); (O.G.-T.); (S.V.)
- Center for Cardiovascular Diseases and Sciences and Center for Redox Biology and Cardiovascular Disease, LSU Health Sciences Center, Shreveport, LA 71103, USA; (X.S.); (H.N.); (A.W.O.); (T.D.); (S.R.B.); (C.G.K.)
| |
Collapse
|
4
|
Pu L, Luo Y, Wen Z, Dai Y, Zheng C, Zhu X, Qin L, Zhang C, Liang H, Zhang J, Guo L, Wang L. GPX2 Gene Affects Feed Efficiency of Pigs by Inhibiting Fat Deposition and Promoting Muscle Development. Animals (Basel) 2022; 12:ani12243528. [PMID: 36552449 PMCID: PMC9774625 DOI: 10.3390/ani12243528] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
GPX2 has been recognized as a potential candidate gene for feed efficiency in pigs. This article aimed to elucidate polymorphism of GPX2 associated with feed efficiency and its related molecular mechanism. In this study, seven single nucleotide polymorphisms (SNP) of GPX2 were found among 383 Duroc pigs. In addition, seven SNPs and ALGA0043483 (PorcineSNP60 BeadChip data in 600 Duroc pigs), which are near the GPX2 gene, were identified in one haplotypes block. Furthermore, associated studies showed that the genotype of GPX2 has significant association with weaning weight and 100 kg BF in Duroc pigs. In addition, the AG had no effect when the backfat became thinner, and the FCR and RFI traits had a tendency to decrease in the G3 + TT combination genotype, accompanied by an increase of GPX2 expression in backfat and muscle tissues. At the cellular level, the adipocyte proliferation and ability of adipogenic differentiation were reduced, and the lipid degradation increased in 3T3-L1 when there was overexpression of GPX2. In contrast, overexpression of the GPX2 gene can promote the muscle cell proliferation and myogenic differentiation in C2C12 cells. In other words, GPX2 has the effect of reducing fat deposition and promoting muscle development, and it is a candidate gene for backfat and feed efficiency.
Collapse
Affiliation(s)
- Lei Pu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Correspondence: (L.P.); (L.W.)
| | - Yunyan Luo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Zuochen Wen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Yuxin Dai
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Chunting Zheng
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Xueli Zhu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Lei Qin
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Chunguang Zhang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Hong Liang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Jianbin Zhang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Liang Guo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
| | - Lixian Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Correspondence: (L.P.); (L.W.)
| |
Collapse
|
5
|
Zager RA, Johnson ACM, Guillem A, Keyser J, Singh B. A Pharmacologic "Stress Test" for Assessing Select Antioxidant Defenses in Patients with CKD. Clin J Am Soc Nephrol 2020; 15:633-642. [PMID: 32291269 PMCID: PMC7269210 DOI: 10.2215/cjn.15951219] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/20/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVES Oxidative stress is a hallmark and mediator of CKD. Diminished antioxidant defenses are thought to be partly responsible. However, there is currently no way to prospectively assess antioxidant defenses in humans. Tin protoporphyrin (SnPP) induces mild, transient oxidant stress in mice, triggering increased expression of select antioxidant proteins (e.g., heme oxygenase 1 [HO-1], NAD[P]H dehydrogenase [quinone] 1 [NQO1], ferritin, p21). Hence, we tested the hypothesis that SnPP can also variably increase these proteins in humans and can thus serve as a pharmacologic "stress test" for gauging gene responsiveness and antioxidant reserves. DESIGN , setting, participants, & measurementsA total of 18 healthy volunteers and 24 participants with stage 3 CKD (n=12; eGFR 30-59 ml/min per 1.73 m2) or stage 4 CKD (n=12; eGFR 15-29 ml/min per 1.73 m2) were injected once with SnPP (9, 27, or 90 mg). Plasma and/or urinary antioxidant proteins were measured at baseline and for up to 4 days post-SnPP dosing. Kidney safety was gauged by serial measurements of BUN, creatinine, eGFR, albuminuria, and four urinary AKI biomarkers (kidney injury molecule 1, neutrophil gelatinase-associated lipocalin, cystatin C, and N-acetyl glucosaminidase). RESULTS Plasma HO-1, ferritin, p21, and NQO1 were all elevated at baseline in CKD participants. Plasma HO-1 and urine NQO1 levels each inversely correlated with eGFR (r=-0.85 to -0.95). All four proteins manifested statistically significant dose- and time-dependent elevations after SnPP injection. However, marked intersubject differences were observed. p21 responses to high-dose SnPP and HO-1 responses to low-dose SnPP were significantly suppressed in participants with CKD versus healthy volunteers. SnPP was well tolerated by all participants, and no evidence of nephrotoxicity was observed. CONCLUSIONS SnPP can be safely administered and, after its injection, the resulting changes in plasma HO-1, NQO1, ferritin, and p21 concentrations can provide information as to antioxidant gene responsiveness/reserves in subjects with and without kidney disease. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER A Study with RBT-1, in Healthy Volunteers and Subjects with Stage 3-4 Chronic Kidney Disease, NCT0363002 and NCT03893799.
Collapse
Affiliation(s)
- Richard A Zager
- Clinical Research Division, The Fred Hutchinson Cancer Research Center, Seattle, Washington .,Department of Medicine, The University of Washington, Seattle, Washington
| | - Ali C M Johnson
- Clinical Research Division, The Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | | - Bhupinder Singh
- Renibus Therapeutics, Southlake, Texas.,Department of Medicine, The University of California, Irvine, California
| |
Collapse
|
6
|
Niyonizigiye I, Ngabire D, Patil MP, Singh AA, Kim GD. In vitro induction of endoplasmic reticulum stress in human cervical adenocarcinoma HeLa cells by fucoidan. Int J Biol Macromol 2019; 137:844-852. [DOI: 10.1016/j.ijbiomac.2019.07.043] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/02/2019] [Accepted: 07/07/2019] [Indexed: 12/24/2022]
|
7
|
Meijles DN, Zoumpoulidou G, Markou T, Rostron KA, Patel R, Lay K, Handa BS, Wong B, Sugden PH, Clerk A. The cardiomyocyte "redox rheostat": Redox signalling via the AMPK-mTOR axis and regulation of gene and protein expression balancing survival and death. J Mol Cell Cardiol 2019; 129:118-129. [PMID: 30771309 PMCID: PMC6497135 DOI: 10.1016/j.yjmcc.2019.02.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/16/2019] [Accepted: 02/12/2019] [Indexed: 02/08/2023]
Abstract
Reactive oxygen species (ROS) play a key role in development of heart failure but, at a cellular level, their effects range from cytoprotection to induction of cell death. Understanding how this is regulated is crucial to develop novel strategies to ameliorate only the detrimental effects. Here, we revisited the fundamental hypothesis that the level of ROS per se is a key factor in the cellular response by applying different concentrations of H2O2 to cardiomyocytes. High concentrations rapidly reduced intracellular ATP and inhibited protein synthesis. This was associated with activation of AMPK which phosphorylated and inhibited Raptor, a crucial component of mTOR complex-1 that regulates protein synthesis. Inhibition of protein synthesis by high concentrations of H2O2 prevents synthesis of immediate early gene products required for downstream gene expression, and such mRNAs (many encoding proteins required to deal with oxidant stress) were only induced by lower concentrations. Lower concentrations of H2O2 promoted mTOR phosphorylation, associated with differential recruitment of some mRNAs to the polysomes for translation. Some of the upregulated genes induced by low H2O2 levels are cytoprotective. We identified p21Cip1/WAF1 as one such protein, and preventing its upregulation enhanced the rate of cardiomyocyte apoptosis. The data support the concept of a "redox rheostat" in which different degrees of ROS influence cell energetics and intracellular signalling pathways to regulate mRNA and protein expression. This sliding scale determines cell fate, modulating survival vs death.
Collapse
Affiliation(s)
- Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London SW17 0RE, UK; School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK.
| | - Georgia Zoumpoulidou
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Thomais Markou
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK; National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Kerry A Rostron
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK
| | - Rishi Patel
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Kenneth Lay
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Balvinder S Handa
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Bethany Wong
- National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Peter H Sugden
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK; National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| | - Angela Clerk
- School of Biological Sciences, University of Reading, Whiteknights, Reading RG6 6AS, UK; National Heart and Lung Institute (Cardiovascular Sciences), Faculty of Medicine, Flowers Building, Imperial College, SW7 2AZ, UK and Dovehouse Street, London SW3 6LY, UK
| |
Collapse
|
8
|
Zobel S, Lorenz M, Frascaroli G, Böhnke J, Bilz NC, Stanifer ML, Boulant S, Bergs S, Liebert UG, Claus C. Rubella Virus Strain-Associated Differences in the Induction of Oxidative Stress Are Independent of Their Interferon Activation. Viruses 2018; 10:v10100540. [PMID: 30282907 PMCID: PMC6213305 DOI: 10.3390/v10100540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/26/2018] [Accepted: 09/28/2018] [Indexed: 01/16/2023] Open
Abstract
Rubella virus (RV) infection impacts cellular metabolic activity in a complex manner with strain-specific nutritional requirements. Here we addressed whether this differential metabolic influence was associated with differences in oxidative stress induction and subsequently with innate immune response activation. The low passaged clinical isolates of RV examined in this study induced oxidative stress as validated through generation of the reactive oxygen species (ROS) cytoplasmic hydrogen peroxide and mitochondrial superoxide. The addition of the cytoplasmic and mitochondrial ROS scavengers N-acetyl-l-cysteine and MitoTEMPO, respectively, reduced RV-associated cytopathogenicity and caspase activation. While the degree of oxidative stress induction varied among RV clinical isolates, the level of innate immune response and interferon-stimulated gene activation was comparable. The type III IFNs were highly upregulated in all cell culture systems tested. However, only pre-stimulation with IFN β slightly reduced RV replication indicating that RV appears to have evolved the ability to counteract innate immune response mechanisms. Through the data presented, we showed that the ability of RV to induce oxidative stress was independent of its capacity to stimulate and counteract the intrinsic innate immune response.
Collapse
Affiliation(s)
- Sarah Zobel
- Institute of Virology, University of Leipzig, 04103 Leipzig, Germany.
| | - Mechthild Lorenz
- Institute of Virology, University of Leipzig, 04103 Leipzig, Germany.
| | - Giada Frascaroli
- Leibniz Institute for Experimental Virology, Heinrich Pette Institute, 20251 Hamburg, Germany.
| | - Janik Böhnke
- Institute of Virology, University of Leipzig, 04103 Leipzig, Germany.
| | - Nicole C Bilz
- Institute of Virology, University of Leipzig, 04103 Leipzig, Germany.
| | - Megan L Stanifer
- Schaller Research Group at CellNetworks, Department of Infectious Diseases, Virology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Steeve Boulant
- Schaller Research Group at CellNetworks, Department of Infectious Diseases, Virology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
- Research Group "Cellular Polarity and Viral Infection" (F140), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Sandra Bergs
- Institute of Virology, University of Leipzig, 04103 Leipzig, Germany.
| | - Uwe G Liebert
- Institute of Virology, University of Leipzig, 04103 Leipzig, Germany.
| | - Claudia Claus
- Institute of Virology, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
9
|
Li H, Zou T, Meng S, Peng YZ, Yang JF. p21 protects cardiomyocytes against ischemia-reperfusion injury by inhibiting oxidative stress. Mol Med Rep 2018; 17:4665-4671. [PMID: 29328456 DOI: 10.3892/mmr.2018.8382] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 01/27/2017] [Indexed: 11/06/2022] Open
Abstract
Ischemic heart disease is a major health threat, resulting in a large number of mortalities annually worldwide. Oxidative stress is one of the main causes of cell death during ischemia‑reperfusion (IR) injury. Cyclin dependent kinase inhibitor 1A (known as p21) is important in protecting tissues against IR injury, however the mechanism remains unknown. In the present study, oxygen‑glucose deprivation and subsequent reoxygenation (OGD/R) in H9c2 heart‑derived myocytes was used as a model to study myocardial IR injury in vitro. mRNA and protein expression levels were determined by reverse transcription‑quantitative polymerase chain reaction and western blotting, respectively. The levels of reactive oxygen species were measured using the fluorescence dye 2',7'‑dichlorodihydrofluorescein diacetate. The present data demonstrated that p21 expression was upregulated by tumor protein p53 (p53) in H9c2 cells exposed to OGD/R. p21 protected H9c2 cells against OGD/R‑induced oxidative stress. In addition, p21 mediated upregulation of NF‑E2‑related factor‑2 (Nrf2), a regulator of antioxidant responses, which in turn suppressed cell death in H9c2 cells subjected to OGD/R. Thus, activation of the p53/p21/Nrf2 signaling pathway may be an important adaptive response that limits oxidative injury during IR.
Collapse
Affiliation(s)
- Hong Li
- Department of Emergency Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Tong Zou
- Department of Cardiology, Beijing Hospital, Beijing 100730, P.R. China
| | - Shuai Meng
- Department of Emergency Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Yun-Zhu Peng
- Department of Cardiology, The First Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Jie-Fu Yang
- Department of Cardiology, Beijing Hospital, Beijing 100730, P.R. China
| |
Collapse
|
10
|
Pham HHT, Seong YA, Ngabire D, Oh CW, Kim GD. Cyperus amuricus induces G1 arrest and apoptosis through endoplasmic reticulum stress and mitochondrial signaling in human hepatocellular carcinoma Hep3B cells. JOURNAL OF ETHNOPHARMACOLOGY 2017; 208:157-164. [PMID: 28684299 DOI: 10.1016/j.jep.2017.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/21/2017] [Accepted: 07/02/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGY RELEVANCE Cyperus amuricus (C. amuricus), belongs to the family Cyperaceae, was used to exert wound healing, diuretic, astringent and other intestinal problems in oriental medicine. However, only a few studies have reported its anticancer activities. AIM OF THE STUDY In this study, we determined the activity of C. amuricus on ER stress-induced cell death and G1 cell cycle arrest in human hepatocellular carcinoma (HCC) Hep3B cells. MATERIALS AND METHODS The in vitro cell proliferation assay of C. amuricus was tested on Hep3B and human embryonic kidney HEK293 cells. Subsequently, the cell cycle distribution in the indicated stages using flow cytometric analysis, the expression of cell cycle-related proteins by western blot analysis and immunofluorescence detection of p21CIP1/WAF1 were determined for the comprehensive identification of cell cycle arrest in Hep3B cells. The effect of C. amuricus on the expression of apoptosis-related proteins in Hep3B cells was also performed by western blot analysis. Furthermore, induction of the ER stress mediators in C. amuricus-treated Hep3B cells were observed by western blot analysis, intracellular Ca2+ mobilization assay and immunofluorescence detection of caspase-12. RESULTS C. amuricus strongly exhibited cytotoxic activity on Hep3B cells, but not on HEK293 cells. C. amuricus affected the phosphorylation levels of endoplasmic reticulum sensors and increased the expression of GRP78/BiP and CHOP, resulting in the accumulation of unfolded proteins in the ER and triggering the unfolded protein response. These changes occurred by the elevation of intracellular Ca2+ concentrations, which contributed to ER stress-induced apoptosis in C. amuricus-treated Hep3B cells. C. amuricus also coordinated the stimulation of ER chaperones, which initiated G1 cell cycle arrest through the induction of CDKIs and the inhibition of cyclins and CDKs. Furthermore, C. amuricus triggered apoptosis through the activation of mitochondrial-dependent pathway in Hep3B cells. CONCLUSIONS Our results suggest that C. amuricus is an effective apoptosis inducing agent for Hep3B cells via the G1 arrest, ER stress and mitochondrial mediated apoptotic pathways.
Collapse
Affiliation(s)
- Hai Ha Thi Pham
- Department of Microbiology, College of Natural Sciences, Pukyong National University, Busan 48513, Republic of Korea
| | - Yeong-Ae Seong
- Department of Microbiology, College of Natural Sciences, Pukyong National University, Busan 48513, Republic of Korea
| | - Daniel Ngabire
- Department of Microbiology, College of Natural Sciences, Pukyong National University, Busan 48513, Republic of Korea
| | - Chul-Woong Oh
- Department of Marine Biology, College of Fisheries Sciences, Pukyong National University, Busan 48513, Republic of Korea
| | - Gun-Do Kim
- Department of Microbiology, College of Natural Sciences, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
11
|
Pujadas G, De Nigris V, Prattichizzo F, La Sala L, Testa R, Ceriello A. The dipeptidyl peptidase-4 (DPP-4) inhibitor teneligliptin functions as antioxidant on human endothelial cells exposed to chronic hyperglycemia and metabolic high-glucose memory. Endocrine 2017; 56:509-520. [PMID: 27530507 PMCID: PMC5435779 DOI: 10.1007/s12020-016-1052-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/04/2016] [Indexed: 12/25/2022]
Abstract
Dipeptidyl peptidase-4 inhibitors are widely used in type 2 diabetes. Endothelium plays a crucial role maintaining vascular integrity and function. Chronic exposure to high glucose drives to endothelial dysfunction generating oxidative stress. Teneligliptin is a novel dipeptidyl peptidase-4 inhibitor with antioxidant properties. This study is aimed to verify a potential protective action of teneligliptin in endothelial cells exposed to high glucose. Human umbilical vein endothelial cells were cultured under normal (5 mmol/L) or high glucose (25 mmol/L) during 21 days, or at high glucose during 14 days followed by 7 days at normal glucose, to reproduce the high-metabolic memory state. During this period, different concentrations of teneligliptin (0.1, 1.0 and 3.0 µmol/L) or sitagliptin (0.5 µmol/L) were added to cells. Ribonucleic acid and protein expression were assessed for antioxidant response, proliferation, apoptosis and endoplasmic reticulum stress markers. Teneligliptin promotes the antioxidant response in human umbilical vein endothelial cells, reducing ROS levels and inducing Nrf2-target genes messenger ribonucleic acid expression. Teneligliptin, but not sitagliptin, reduces the expression of the nicotine amide adenine dinucleotide phosphate oxidase regulatory subunit P22 -phox , however, both blunt the high glucose-induced increase of TXNIP. Teneligliptin improves proliferation rates in human umbilical vein endothelial cells exposed to high glucose, regulating the expression of cell-cycle inhibitors markers (P53, P21 and P27), and reducing proapoptotic genes (BAX and CASP3), while promotes BCL2 expression. Teneligliptin ameliorates high glucose-induced endoplasmic reticulum stress reducing the expression of several markers (BIP, PERK, ATF4, CHOP, IRE1a and ATF6). Teneligliptin has antioxidant properties, ameliorates oxidative stress and apoptotic phenotype and it can overcome the metabolic memory effect, induced by chronic exposure to high glucose in human endothelial cells.
Collapse
Affiliation(s)
- Gemma Pujadas
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Hospital Clínic, C/Rosselló, 149-153, 08036, Barcelona, Spain.
| | - Valeria De Nigris
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Hospital Clínic, C/Rosselló, 149-153, 08036, Barcelona, Spain
| | - Francesco Prattichizzo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Hospital Clínic, C/Rosselló, 149-153, 08036, Barcelona, Spain
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Lucia La Sala
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Hospital Clínic, C/Rosselló, 149-153, 08036, Barcelona, Spain
| | - Roberto Testa
- Experimental Models in Clinical Pathology, INRCA-IRCCS National Institute, Ancona, Italy
| | - Antonio Ceriello
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Hospital Clínic, C/Rosselló, 149-153, 08036, Barcelona, Spain.
- IRCCS MultiMedica, Milan, Italy.
| |
Collapse
|
12
|
Forred BJ, Daugaard DR, Titus BK, Wood RR, Floen MJ, Booze ML, Vitiello PF. Detoxification of Mitochondrial Oxidants and Apoptotic Signaling Are Facilitated by Thioredoxin-2 and Peroxiredoxin-3 during Hyperoxic Injury. PLoS One 2017; 12:e0168777. [PMID: 28045936 PMCID: PMC5207683 DOI: 10.1371/journal.pone.0168777] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 12/06/2016] [Indexed: 01/22/2023] Open
Abstract
Mitochondria play a fundamental role in the regulation of cell death during accumulation of oxidants. High concentrations of atmospheric oxygen (hyperoxia), used clinically to treat tissue hypoxia in premature newborns, is known to elicit oxidative stress and mitochondrial injury to pulmonary epithelial cells. A consequence of oxidative stress in mitochondria is the accumulation of peroxides which are detoxified by the dedicated mitochondrial thioredoxin system. This system is comprised of the oxidoreductase activities of peroxiredoxin-3 (Prx3), thioredoxin-2 (Trx2), and thioredoxin reductase-2 (TrxR2). The goal of this study was to understand the role of the mitochondrial thioredoxin system and mitochondrial injuries during hyperoxic exposure. Flow analysis of the redox-sensitive, mitochondrial-specific fluorophore, MitoSOX, indicated increased levels of mitochondrial oxidant formation in human adenocarcinoma cells cultured in 95% oxygen. Increased expression of Trx2 and TrxR2 in response to hyperoxia were not attributable to changes in mitochondrial mass, suggesting that hyperoxic upregulation of mitochondrial thioredoxins prevents accumulation of oxidized Prx3. Mitochondrial oxidoreductase activities were modulated through pharmacological inhibition of TrxR2 with auranofin and genetically through shRNA knockdown of Trx2 and Prx3. Diminished Trx2 and Prx3 expression was associated with accumulation of mitochondrial superoxide; however, only shRNA knockdown of Trx2 increased susceptibility to hyperoxic cell death and increased phosphorylation of apoptosis signal-regulating kinase-1 (ASK1). In conclusion, the mitochondrial thioredoxin system regulates hyperoxic-mediated death of pulmonary epithelial cells through detoxification of oxidants and regulation of redox-dependent apoptotic signaling.
Collapse
Affiliation(s)
- Benjamin J. Forred
- Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Darwin R. Daugaard
- Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Brianna K. Titus
- Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Ryan R. Wood
- Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Miranda J. Floen
- Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Michelle L. Booze
- Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Peter F. Vitiello
- Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, South Dakota, United States of America
| |
Collapse
|
13
|
Fielder E, von Zglinicki T, Jurk D. The DNA Damage Response in Neurons: Die by Apoptosis or Survive in a Senescence-Like State? J Alzheimers Dis 2017; 60:S107-S131. [PMID: 28436392 DOI: 10.3233/jad-161221] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurons are exposed to high levels of DNA damage from both physiological and pathological sources. Neurons are post-mitotic and their loss cannot be easily recovered from; to cope with DNA damage a complex pathway called the DNA damage response (DDR) has evolved. This recognizes the damage, and through kinases such as ataxia-telangiectasia mutated (ATM) recruits and activates downstream factors that mediate either apoptosis or survival. This choice between these opposing outcomes integrates many inputs primarily through a number of key cross-road proteins, including ATM, p53, and p21. Evidence of re-entry into the cell-cycle by neurons can be seen in aging and diseases such as Alzheimer's disease. This aberrant cell-cycle re-entry is lethal and can lead to the apoptotic death of the neuron. Many downstream factors of the DDR promote cell-cycle arrest in response to damage and appear to protect neurons from apoptotic death. However, neurons surviving with a persistently activated DDR show all the features known from cell senescence; including metabolic dysregulation, mitochondrial dysfunction, and the hyper-production of pro-oxidant, pro-inflammatory and matrix-remodeling factors. These cells, termed senescence-like neurons, can negatively influence the extracellular environment and may promote induction of the same phenotype in surrounding cells, as well as driving aging and age-related diseases. Recently developed interventions targeting the DDR and/or the senescent phenotype in a range of non-neuronal tissues are being reviewed as they might become of therapeutic interest in neurodegenerative diseases.
Collapse
Affiliation(s)
- Edward Fielder
- The Ageing Biology Centre and Institute for Cell and Molecular Biology, Newcastle University, Newcastle Upon Tyne, UK
| | - Thomas von Zglinicki
- The Ageing Biology Centre and Institute for Cell and Molecular Biology, Newcastle University, Newcastle Upon Tyne, UK
| | - Diana Jurk
- The Ageing Biology Centre and Institute for Cell and Molecular Biology, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
14
|
P21Waf1/Cip1 plays a critical role in furazolidone-induced apoptosis in HepG2 cells through influencing the caspase-3 activation and ROS generation. Food Chem Toxicol 2016; 88:1-12. [DOI: 10.1016/j.fct.2015.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/08/2015] [Accepted: 12/03/2015] [Indexed: 12/14/2022]
|
15
|
Abstract
Oxygen is the basic molecule which supports life and it truly is "god's gift to life." Despite its immense importance, research on "oxygen biology" has never received the light of the day and has been limited to physiological and biochemical studies. It seems that in modern day biology, oxygen research is summarized in one word "hypoxia." Scientists have focused on hypoxia-induced transcriptomics and molecular-cellular alterations exclusively in disease models. Interestingly, the potential of oxygen to control the basic principles of biology like homeostatic maintenance, transcription, replication, and protein folding among many others, at the molecular level, has been completely ignored. Here, we present a perspective on the crucial role played by oxygen in regulation of basic biological phenomena. Our conclusion highlights the importance of establishing novel research areas like oxygen biology, as there is great potential in this field for basic science discoveries and clinical benefits to the society.
Collapse
|
16
|
Kondrikov D, Fulton D, Dong Z, Su Y. Heat Shock Protein 70 Prevents Hyperoxia-Induced Disruption of Lung Endothelial Barrier via Caspase-Dependent and AIF-Dependent Pathways. PLoS One 2015; 10:e0129343. [PMID: 26066050 PMCID: PMC4465980 DOI: 10.1371/journal.pone.0129343] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 05/07/2015] [Indexed: 12/03/2022] Open
Abstract
Exposure of pulmonary artery endothelial cells (PAECs) to hyperoxia results in a compromise in endothelial monolayer integrity, an increase in caspase-3 activity, and nuclear translocation of apoptosis-inducing factor (AIF), a marker of caspase-independent apoptosis. In an endeavor to identify proteins involved in hyperoxic endothelial injury, we found that the protein expression of heat-shock protein 70 (Hsp70) was increased in hyperoxic PAECs. The hyperoxia-induced Hsp70 protein expression is from hspA1B gene. Neither inhibition nor overexpression of Hsp70 affected the first phase barrier disruption of endothelial monolayer. Nevertheless, inhibition of Hsp70 by using the Hsp70 inhibitor KNK437 or knock down Hsp70 using siRNA exaggerated and overexpression of Hsp70 prevented the second phase disruption of lung endothelial integrity. Moreover, inhibition of Hsp70 exacerbated and overexpression of Hsp70 prevented hyperoxia-induced apoptosis, caspase-3 activation, and increase in nuclear AIF protein level in PAECs. Furthermore, we found that Hsp70 interacted with AIF in the cytosol in hyperoxic PAECs. Inhibition of Hsp70/AIF association by KNK437 correlated with increased nuclear AIF level and apoptosis in KNK437-treated PAECs. Finally, the ROS scavenger NAC prevented the hyperoxia-induced increase in Hsp70 expression and reduced the interaction of Hsp70 with AIF in hyperoxic PAECs. Together, these data indicate that increased expression of Hsp70 plays a protective role against hyperoxia-induced lung endothelial barrier disruption through caspase-dependent and AIF-dependent apoptotic pathways. Association of Hsp70 with AIF prevents AIF nuclear translocation, contributing to the protective effect of Hsp70 on hyperoxia-induced endothelial apoptosis. The hyperoxia-induced increase in Hsp70 expression and Hsp70/AIF interaction is contributed to ROS formation.
Collapse
Affiliation(s)
- Dmitry Kondrikov
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, United States of America
| | - David Fulton
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, United States of America
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, United States of America
| | - Zheng Dong
- Department of Cell Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, United States of America
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia 30912, United States of America
| | - Yunchao Su
- Department of Pharmacology & Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, United States of America
- Department of Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, United States of America
- Vascular Biology Center, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, United States of America
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia 30912, United States of America
- * E-mail:
| |
Collapse
|
17
|
Burton DGA, Faragher RGA. Cellular senescence: from growth arrest to immunogenic conversion. AGE (DORDRECHT, NETHERLANDS) 2015; 37:27. [PMID: 25787341 PMCID: PMC4365077 DOI: 10.1007/s11357-015-9764-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 02/27/2015] [Indexed: 05/23/2023]
Abstract
Cellular senescence was first reported in human fibroblasts as a state of stable in vitro growth arrest following extended culture. Since that initial observation, a variety of other phenotypic characteristics have been shown to co-associate with irreversible cell cycle exit in senescent fibroblasts. These include (1) a pro-inflammatory secretory response, (2) the up-regulation of immune ligands, (3) altered responses to apoptotic stimuli and (4) promiscuous gene expression (stochastic activation of genes possibly as a result of chromatin remodeling). Many features associated with senescent fibroblasts appear to promote conversion to an immunogenic phenotype that facilitates self-elimination by the immune system. Pro-inflammatory cytokines can attract and activate immune cells, the presentation of membrane bound immune ligands allows for specific recognition and promiscuous gene expression may function to generate an array of tissue restricted proteins that could subsequently be processed into peptides for presentation via MHC molecules. However, the phenotypes of senescent cells from different tissues and species are often assumed to be broadly similar to those seen in senescent human fibroblasts, but the data show a more complex picture in which the growth arrest mechanism, tissue of origin and species can all radically modulate this basic pattern. Furthermore, well-established triggers of cell senescence are often associated with a DNA damage response (DDR), but this may not be a universal feature of senescent cells. As such, we discuss the role of DNA damage in regulating an immunogenic response in senescent cells, in addition to discussing less established "atypical" senescent states that may occur independent of DNA damage.
Collapse
Affiliation(s)
- D. G. A. Burton
- Department of Molecular Cell Biology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | - R. G. A. Faragher
- School of Pharmacy & Biomolecular Science, University of Brighton, Huxley Building, Brighton, UK
| |
Collapse
|
18
|
Li J, Chen Y, Wan J, Liu X, Yu C, Li W. ABT-263 enhances sorafenib-induced apoptosis associated with Akt activity and the expression of Bax and p21((CIP1/WAF1)) in human cancer cells. Br J Pharmacol 2015; 171:3182-95. [PMID: 24571452 DOI: 10.1111/bph.12659] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 02/16/2014] [Accepted: 02/21/2014] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Sorafenib, a potent inhibitor that targets several kinases associated with tumourigenesis and cell survival, has been approved for clinical treatment as a single agent. However, combining sorafenib with other agents improves its anti-tumour efficacy in various preclinical tumour models. ABT-263, a second-generation BH3 mimic, binds to the anti-apoptotic family members Bcl-2, Bcl-xL and Bcl-w, and has been demonstrated to enhance TNFSF10 (TRAIL)-induced apoptosis in human hepatocarcinoma cells. Hence, we investigated the effects of ABT-263 treatment combined with sorafenib. EXPERIMENTAL APPROACH The effects of ABT-263 combined with sorafenib were investigated in vitro, on cell viability, clone formation and apoptosis, and the mechanism examined using western blot and flow cytometry. This combination was also evaluated in vivo, in a mouse xenograft model; tumour growth, volume and weights were measured and a TUNEL assay performed. KEY RESULTS ABT-263 enhanced sorafenib-induced apoptosis while sparing non-tumourigenic cells. Although ABT-263 plus sorafenib significantly stimulated intracellular reactive oxygen species production and subsequent mitochondrial depolarization, this was not sufficient to trigger cell apoptosis. ABT-263 plus sorafenib significantly decreased Akt activity, which was, at least partly, involved in its effect on apoptosis. Bax and p21 (CIP1/WAF1) were shown to play a critical role in ABT-263 plus sorafenib-induced apoptosis. Combining sorafenib with ABT-263 dramatically increased its efficacy in vivo. CONCLUSION AND IMPLICATIONS The anti-tumour activity of ABT-263 plus sorafenib may involve the induction of intrinsic cell apoptosis via inhibition of Akt, and reduced Bax and p21 expression. Our findings offer a novel effective therapeutic strategy for tumour treatment.
Collapse
Affiliation(s)
- Jingru Li
- College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | | | | | | | | | | |
Collapse
|
19
|
Zhu J, Chen M, Chen N, Ma A, Zhu C, Zhao R, Jiang M, Zhou J, Ye L, Fu H, Zhang X. Glycyrrhetinic acid induces G1‑phase cell cycle arrest in human non‑small cell lung cancer cells through endoplasmic reticulum stress pathway. Int J Oncol 2015; 46:981-8. [PMID: 25573651 PMCID: PMC4324580 DOI: 10.3892/ijo.2015.2819] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/20/2014] [Indexed: 12/13/2022] Open
Abstract
Glycyrrhetinic acid (GA) is a natural compound extracted from liquorice, which is often used in traditional Chinese medicine. The purpose of the present study was to investigate the antitumor effect of GA in human non-small cell lung cancer (NSCLC), and its underlying mechanisms in vitro. We have shown that GA suppressed the proliferation of A549 and NCI-H460 cells. Flow cytometric analysis showed that GA arrested cell cycle in G0/G1 phase without inducing apoptosis. Western blot analysis indicated that GA mediated G1-phase cell cycle arrest by upregulation of cyclin-dependent kinase inhibitors (CKIs) (p18, p16, p27 and p21) and inhibition of cyclins (cyclin-D1, -D3 and -E) and cyclin-dependent kinases (CDKs) (CDK4, 6 and 2). GA also maintained pRb phosphorylation status, and inhibited E2F transcription factor 1 (E2F-1) in both cell lines. GA upregulated the unfolded proteins, Bip, PERK and ERP72. Accumulation of unfolded proteins in the endoplasmic reticulum (ER) triggered the unfolded protein response (UPR), which could be the mechanism by which GA inhibited cell proliferation in NSCLC cells. GA then coordinated the induction of ER chaperones, which decreased protein synthesis and induced cell cycle arrest in the G1 phase. This study provides experimental evidence to support the development of GA as a chemotherapeutic agent for NSCLC.
Collapse
Affiliation(s)
- Jie Zhu
- The First Clinical Medicine College, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Meijuan Chen
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Ning Chen
- The First Clinical Medicine College, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Aizhen Ma
- The First Clinical Medicine College, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Chunyan Zhu
- The First Clinical Medicine College, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Ruolin Zhao
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Miao Jiang
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Jing Zhou
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Lihong Ye
- The First Clinical Medicine College, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Haian Fu
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xu Zhang
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| |
Collapse
|
20
|
Biology of the cell cycle inhibitor p21CDKN1A: molecular mechanisms and relevance in chemical toxicology. Arch Toxicol 2014; 89:155-78. [DOI: 10.1007/s00204-014-1430-4] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 12/03/2014] [Indexed: 02/07/2023]
|
21
|
Ko F, Isoda F, Mobbs C. Laparotomy in mice induces blood cell expression of inflammatory and stress genes. J Interferon Cytokine Res 2014; 35:302-12. [PMID: 25406893 DOI: 10.1089/jir.2014.0031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Surgical trauma induces immune and stress responses although its effects on postsurgical inflammatory and stress gene expression remain poorly characterized. This study sought to improve current scientific knowledge by investigating the effects of laparotomy on mouse blood cell inflammatory and stress gene expression. Three-month-old male C57BL/6J mice were subjected to 2% isoflurane or 2% isoflurane with laparotomy and sacrificed 4 h postintervention. Blood was collected and blood cell expression of 158 genes central to inflammatory and stress responses was assayed using quantitative polymerase chain reaction arrays. Mice subjected to isoflurane with laparotomy, compared with mice receiving isoflurane alone, had >2-fold upregulation of genes in inflammation (Osm, IL1rn, IL1b, and Csf1), oxidative stress (Hmox1), heat shock (Hspa1b), growth arrest (Cdkn1a), and DNA repair (Ugt1a2). These genes demonstrated similar expression patterns by Pearson correlation and cluster analysis. Thus, laparotomy induces coordinated, postsurgical blood cell expression of unique inflammatory and stress genes whose roles in influencing surgical outcomes need further investigation.
Collapse
Affiliation(s)
- Fred Ko
- 1 Brookdale Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai , New York, New York
| | | | | |
Collapse
|
22
|
Zhang P, Suidasari S, Hasegawa T, Yanaka N, Kato N. Vitamin B₆ activates p53 and elevates p21 gene expression in cancer cells and the mouse colon. Oncol Rep 2014; 31:2371-6. [PMID: 24626782 DOI: 10.3892/or.2014.3073] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 02/10/2014] [Indexed: 11/06/2022] Open
Abstract
Increasing evidence indicates vitamin B6 acts as a protective factor against colon cancer. However, the mechanisms of the effect of vitamin B6 are poorly understood. The present preliminary study using DNA microarray and real-time PCR indicates p21 mRNA is upregulated in human colon carcinoma (HT29) cells exposed to pyridoxal (PL, 500 µM). A similar effect was observed in human epithelial colorectal adenocarcinoma (Caco2) cells, human colon adenocarcinoma (LoVo) cells, human embryonic kidney (HEK293T) cells, and human hepatoma (HepG2) cells. Adding other B6-vitamers such as pyridoxal 5'-phosphate (PLP), pyridoxine (PN), and pyridoxamine (PM) caused no such effect. In order to understand the mechanism of higher mRNA expression of p21 by PL, effect of PL on the p53 activation was examined (the upstream factor for p21 mRNA transcription) in HT29 cells, LoVo cells, and HepG2 cells. PL increased the phosphorylated p53 protein levels (active form) in whole-cell lysates and the nuclei of the cells. Noteworthy, the consumption of a vitamin B6-deficient diet for 5 weeks significantly reduced p21 mRNA levels and tended to reduce phosphorylated p53 protein levels (P=0.053) in the colons of mice compared to a diet with adequate vitamin B6. Thus, these results suggest vitamin B6 plays a role in increasing p21 gene expression via p53 activation in several cancer cells and the mouse colon.
Collapse
Affiliation(s)
- Peipei Zhang
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Sofya Suidasari
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Tomomi Hasegawa
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Noriyuki Yanaka
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| | - Norihisa Kato
- Graduate School of Biosphere Science, Hiroshima University, Higashi-Hiroshima 739-8528, Japan
| |
Collapse
|
23
|
Hernandez AM, Colvin ES, Chen YC, Geiss SL, Eller LE, Fueger PT. Upregulation of p21 activates the intrinsic apoptotic pathway in β-cells. Am J Physiol Endocrinol Metab 2013; 304:E1281-90. [PMID: 23592481 PMCID: PMC3680698 DOI: 10.1152/ajpendo.00663.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Diabetes manifests from a loss in functional β-cell mass, which is regulated by a dynamic balance of various cellular processes, including β-cell growth, proliferation, and death as well as secretory function. The cell cycle machinery comprised of cyclins, kinases, and inhibitors regulates proliferation. However, their involvement during β-cell stress during the development of diabetes is not well understood. Interestingly, in a screen of multiple cell cycle inhibitors, p21 was dramatically upregulated in INS-1-derived 832/13 cells and rodent islets by two pharmacological inducers of β-cell stress, dexamethasone and thapsigargin. We hypothesized that β-cell stress upregulates p21 to activate the apoptotic pathway and suppress cell survival signaling. To this end, p21 was adenovirally overexpressed in pancreatic rat islets and 832/13 cells. As expected, p21 overexpression resulted in decreased [(3)H]thymidine incorporation. Flow cytometry analysis in p21-transduced 832/13 cells verified lower replication, as indicated by a decreased cell population in the S phase and a block in G2/M transition. The sub-G0 cell population was higher with p21 overexpression and was attributable to apoptosis, as demonstrated by increased annexin-positive stained cells and cleaved caspase-3 protein. p21-mediated caspase-3 cleavage was inhibited by either overexpression of the antiapoptotic mitochondrial protein Bcl-2 or siRNA-mediated suppression of the proapoptotic proteins Bax and Bak. Therefore, an intact intrinsic apoptotic pathway is central for p21-mediated cell death. In summary, our findings indicate that β-cell apoptosis can be triggered by p21 during stress and is thus a potential target to inhibit for protection of functional β-cell mass.
Collapse
|
24
|
Hsp90 Inhibitors NVP-AUY922 and NVP-BEP800 May Exert a Significant Radiosensitization on Tumor Cells along with a Cell Type-Specific Cytotoxicity. Transl Oncol 2012; 5:356-69. [PMID: 23066444 DOI: 10.1593/tlo.12211] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 06/28/2012] [Accepted: 07/04/2012] [Indexed: 02/04/2023] Open
Abstract
Targeting heat shock protein 90 (Hsp90) provides a promising therapeutic approach to enhance the sensitivity of tumor cells to ionizing radiation (IR). To explore the impact of scheduling drug-IR administration, in the present study, we analyzed the response of lung carcinoma A549 and glioblastoma SNB19 cells to simultaneous drug-IR treatment followed by a long-term drug administration. Cellular response was evaluated at different time intervals after IR-alone, drug-alone, or combined drug-IR treatments by colony counts and expression profiles of Hsp90 and its clients, along with several apoptotic markers and cell cycle-related proteins, as well as by IR-drug-induced cell cycle arrest, DNA damage, and repair. A short 30-minute exposure to either Hsp90 inhibitor did not affect the radiosensitivity of both tumor cell lines. Increasing the duration of post-IR-drug treatment progressively enhanced the sensitivity of SNB19 cells to IR. In contrast, the response of A549 cells to drug-IR combination was largely determined by the cytotoxic effects of both drugs without radiosensitization. Combined drug-IR treatment induced more severe DNA damage in both tumor cell lines than each treatment alone and also protracted the kinetics of DNA damage repair in SNB19 cells. In addition to large cell cycle disturbances, drug-IR treatment also caused depletion of the antiapoptotic proteins Akt and Raf-1 in both cell lines, along with a decrease of survivin in A549 cells in case of NVP-AUY922. The data show that simultaneous Hsp90 inhibition and irradiation may induce cell type-specific radiosensitization as well as cytotoxicity against tumor cells.
Collapse
|
25
|
Farrow KN, Lee KJ, Perez M, Schriewer JM, Wedgwood S, Lakshminrusimha S, Smith CL, Steinhorn RH, Schumacker PT. Brief hyperoxia increases mitochondrial oxidation and increases phosphodiesterase 5 activity in fetal pulmonary artery smooth muscle cells. Antioxid Redox Signal 2012; 17:460-70. [PMID: 22229392 PMCID: PMC3365357 DOI: 10.1089/ars.2011.4184] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
AIMS Oxygen is a pulmonary vasodilator, but data suggest high O(2) concentrations impede that response. We previously reported 24 h of 100% O(2) increased phosphodiesterase 5 (PDE5) activity in fetal pulmonary artery smooth muscle cells (FPASMC) and in ventilated neonatal lambs. PDE5 degrades cyclic GMP (cGMP) and inhibits nitric oxide (NO)-mediated cGMP-dependent vasorelaxation. We sought to determine the mechanism by which hyperoxia initiates reactive oxygen species (ROS) production and regulates PDE5. RESULTS Thirty minutes of hyperoxia increased mitochondrial ROS versus normoxia (30.3±1.7% vs. 21.1±2.8%), but had no effect on cytosolic ROS, measured by roGFP, a ratiometric protein thiol redox sensor. Hyperoxia increased PDE5 activity (220±39%) and decreased cGMP responsiveness to NO (37±17%). Mitochondrial catalase overexpression attenuated hyperoxia-induced mitochondrial roGFP oxidation, compared to FPASMC infected with empty adenoviral vector (50±3% of control) or mitochondrial superoxide dismutase. MitoTEMPO, mitochondrial catalase, and DT-3, a cGMP-dependent protein kinase I alpha inhibitor, decreased PDE5 activity (32±13%, 26±21%, and 63±10% of control, respectively), and restored cGMP responsiveness to NO (147±16%,172±29%, and 189±43% of control, respectively). C57Bl6 mice exposed to 90%-100% O(2) for 45 min±mechanical ventilation had increased PA PDE5 activity (206±39% and 235±75%, respectively). INNOVATION This is the first description that hyperoxia induces ROS in the mitochondrial matrix prior to the cytosol. Our results indicate that short hyperoxia exposures can produce significant changes in critical cellular signaling pathways. CONCLUSIONS These results indicate that mitochondrial matrix oxidant signals generated during hyperoxia, specifically H(2)O(2), activate PDE5 in a cGMP-dependent protein kinase-dependent manner in pulmonary vascular smooth muscle cells.
Collapse
Affiliation(s)
- Kathryn N Farrow
- Department of Pediatrics, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Vurusaner B, Poli G, Basaga H. Tumor suppressor genes and ROS: complex networks of interactions. Free Radic Biol Med 2012; 52:7-18. [PMID: 22019631 DOI: 10.1016/j.freeradbiomed.2011.09.035] [Citation(s) in RCA: 223] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 09/27/2011] [Accepted: 09/29/2011] [Indexed: 12/24/2022]
Abstract
Tumor suppressor genes regulate diverse cellular activities including DNA damage repair, cell cycle arrest, mitogenic signaling, cell differentiation, migration, and programmed cell death. In this review the tumor suppressor genes p53, FoxO, retinoblastoma (RB), p21, p16, and breast cancer susceptibility genes 1 and 2 (BRCA1 and BRCA2) and their roles in oxidative stress are summarized with a focus on the links and interplay between their pathways and reactive oxygen species (ROS). The results of a number of studies have demonstrated an antioxidant role for tumor suppressor proteins, activating the expression of some well-known antioxidant genes in response to oxidative stress. On the other hand, recent studies have revealed a pro-oxidant role for p53 by which cellular ROS are increased by enhanced transcription of proapoptotic genes. A tightly regulated feedback loop between ROS and FoxO proteins, with ROS regulating FoxO activity through posttranslational modifications and protein interactions and FoxO controlling intracellular ROS levels, has been demonstrated. Furthermore, these studies have shown that FoxO transcription factors and p38 mitogen-activated protein kinases may interact with the RB pathway under stress conditions. In addition, cellular senescence studies established an unexpected role for ROS in inducing and maintaining senescence-induced tumor suppression that blocks cytokinesis to ensure senescent cells never divide again. p21 and p16 have been shown to act as tumor suppressor proteins and this function extends beyond cell cycle control and includes important roles in regulating oxidative stress. Consequently, these important interactions indicate a critical potential role for tumor suppressor genes in the cellular response against oxidative stress and emphasize links between ROS and tumor suppressor genes that might be therapeutic targets in oxidative damage-associated diseases.
Collapse
Affiliation(s)
- Beyza Vurusaner
- Biological Sciences and Bioengineering Program, Faculty of Natural Sciences and Engineering, Sabanci University, Istanbul, Turkey
| | | | | |
Collapse
|
27
|
Costa RO, Ferreiro E, Martins I, Santana I, Cardoso SM, Oliveira CR, Pereira CMF. Amyloid β-induced ER stress is enhanced under mitochondrial dysfunction conditions. Neurobiol Aging 2011; 33:824.e5-16. [PMID: 21704433 DOI: 10.1016/j.neurobiolaging.2011.04.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 04/13/2011] [Accepted: 04/29/2011] [Indexed: 01/10/2023]
Abstract
Previously we reported that endoplasmic reticulum (ER)-mitochondria crosstalk is involved in amyloid-β (Aβ)-induced apoptosis. Now we show that mitochondrial dysfunction affects the ER stress response triggered by Aβ using cybrids that recreate the defect in mitochondrial cytochrome c oxidase (COX) activity detected in platelets from Alzheimer's disease (AD) patients. AD and control cybrids were treated with Aβ or classical ER stressors and the ER stress-mediated apoptotic cell death pathway was accessed. Upon treatment, we found increased glucose-regulated protein 78 (GRP78) levels and caspase-4 activation (ER stress markers) which were more pronounced in AD cybrids. Treated AD cybrids also exhibited decreased cell survival as well as increased caspase-3-like activity, poli-ADP-ribose-polymerase (PARP) levels and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive apoptotic cells. Finally, we showed that Aβ-induced caspase-3 activation in both cybrid cell lines was prevented by dantrolene, thus implicating ER Ca(2+) release in ER stress-mediated apoptosis. Our results demonstrate that mitochondrial dysfunction occurring in AD patients due to COX inhibition potentiates cell susceptibility to Aβ-induced ER stress. This study further supports the close communication between ER and mitochondria during apoptosis in AD.
Collapse
Affiliation(s)
- Rui O Costa
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | | | | | | | | | | |
Collapse
|
28
|
Budinger GRS, Mutlu GM, Urich D, Soberanes S, Buccellato LJ, Hawkins K, Chiarella SE, Radigan KA, Eisenbart J, Agrawal H, Berkelhamer S, Hekimi S, Zhang J, Perlman H, Schumacker PT, Jain M, Chandel NS. Epithelial cell death is an important contributor to oxidant-mediated acute lung injury. Am J Respir Crit Care Med 2011; 183:1043-54. [PMID: 20959557 PMCID: PMC3086743 DOI: 10.1164/rccm.201002-0181oc] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 10/15/2010] [Indexed: 01/11/2023] Open
Abstract
RATIONALE Acute lung injury and the acute respiratory distress syndrome are characterized by increased lung oxidant stress and apoptotic cell death. The contribution of epithelial cell apoptosis to the development of lung injury is unknown. OBJECTIVES To determine whether oxidant-mediated activation of the intrinsic or extrinsic apoptotic pathway contributes to the development of acute lung injury. METHODS Exposure of tissue-specific or global knockout mice or cells lacking critical components of the apoptotic pathway to hyperoxia, a well-established mouse model of oxidant-induced lung injury, for measurement of cell death, lung injury, and survival. MEASUREMENTS AND MAIN RESULTS We found that the overexpression of SOD2 prevents hyperoxia-induced BAX activation and cell death in primary alveolar epithelial cells and prolongs the survival of mice exposed to hyperoxia. The conditional loss of BAX and BAK in the lung epithelium prevented hyperoxia-induced cell death in alveolar epithelial cells, ameliorated hyperoxia-induced lung injury, and prolonged survival in mice. By contrast, Cyclophilin D-deficient mice were not protected from hyperoxia, indicating that opening of the mitochondrial permeability transition pore is dispensable for hyperoxia-induced lung injury. Mice globally deficient in the BH3-only proteins BIM, BID, PUMA, or NOXA, which are proximal upstream regulators of BAX and BAK, were not protected against hyperoxia-induced lung injury suggesting redundancy of these proteins in the activation of BAX or BAK. CONCLUSIONS Mitochondrial oxidant generation initiates BAX- or BAK-dependent alveolar epithelial cell death, which contributes to hyperoxia-induced lung injury.
Collapse
Affiliation(s)
- G. R. Scott Budinger
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Gökhan M. Mutlu
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Daniela Urich
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Saul Soberanes
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Leonard J. Buccellato
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Keenan Hawkins
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sergio E. Chiarella
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Kathryn A. Radigan
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - James Eisenbart
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Hemant Agrawal
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sara Berkelhamer
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Siegfried Hekimi
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jianke Zhang
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Harris Perlman
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Paul T. Schumacker
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Manu Jain
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Navdeep S. Chandel
- Department of Medicine, Department of Cell and Molecular Biology, and Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biology, McGill University, Montreal, Quebec, Canada; and Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Intracellular GTP level determines cell's fate toward differentiation and apoptosis. Toxicol Appl Pharmacol 2011; 253:188-96. [PMID: 21396949 DOI: 10.1016/j.taap.2011.02.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 02/09/2011] [Accepted: 02/28/2011] [Indexed: 01/23/2023]
Abstract
Since the adequate supply of guanine nucleotides is vital for cellular activities, limitation of their syntheses would certainly result in modulation of cellular fate toward differentiation and apoptosis. The aim of this study was to set a correlation between the intracellular level of GTP and the induction of relevant signaling pathways involved in the cell's fate toward life or death. In that regard, we measured the GTP level among human leukemia K562 cells exposed to mycophenolic acid (MPA) or 3-hydrogenkwadaphnin (3-HK) as two potent inosine monophosphate dehydrogenase inhibitors. Our results supported the maturation of the cells when the intracellular GTP level was reduced by almost 30-40%. Under these conditions, 3-HK and/or MPA caused up-regulation of PKCα and PI3K/AKT pathways. Furthermore, co-treatment of cells with hypoxanthine plus 3-HK or MPA, which caused a reduction of about 60% in the intracellular GTP levels, led to apoptosis and activation of mitochondrial pathways through inverse regulation of Bcl-2/Bax expression and activation of caspase-3. Moreover, our results demonstrated that attenuation of GTP by almost 60% augmented the intracellular ROS and nuclear localization of p21 and subsequently led to cell death. These results suggest that two different threshold levels of GTP are needed for induction of differentiation and/or ROS-associated apoptosis.
Collapse
|
30
|
Wu YCM, O'Reilly MA. Bcl-X(L) is the primary mediator of p21 protection against hyperoxia-induced cell death. Exp Lung Res 2010; 37:82-91. [PMID: 21128858 DOI: 10.3109/01902148.2010.521617] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A tight balance between anti- and proapoptotic members of the Bcl-2 family controls cell survival and death. Exposure to hyperoxia shifts this balance towards a prodeath state that ultimately activates Bak- and Bax-dependent cell death. Mechanisms underlying this shift are undefined; however, the cell cycle inhibitor p21 delays the loss of antiapoptotic Mcl-1 and Bcl-X(L), and protects against hyperoxia. Here, H1299 human lung adenocarcinoma cells are used to investigate how these and other members of the Bcl-2 family cooperate with p21 to protect against hyperoxia. Expression of antiapoptotic Mcl-1 and Bcl-X(L), but not Bcl-2 or A1, declined during hyperoxia, whereas proapoptotic Bak, but not Bax, increased. Conditional overexpression of p21 selectively delayed the loss of Mcl-1 and Bcl-X(L), without affecting expression of the other members. siRNA knockdown of Mcl-1 and Bcl-X(L) sensitized cells to hyperoxia, but only the loss of Bcl-X(L) ablated the protective effects of p21. Conversely, overexpression of Mcl-1 and Bcl-X(L) protected against hyperoxia, but only Bcl-X(L) bound Bak and Bax. Altogether, these data suggest that Bcl-X(L) is the primary mediator by which p21 protects against hyperoxia-induced Bak/Bax-dependent cell death.
Collapse
Affiliation(s)
- Yu-Chieh M Wu
- Department of Biomedical Genetics, School of Medicine and Dentistry, The University of Rochester, Rochester, New York 14642, USA
| | | |
Collapse
|
31
|
Verbeke S, Meignan S, Lagadec C, Germain E, Hondermarck H, Adriaenssens E, Le Bourhis X. Overexpression of p75(NTR) increases survival of breast cancer cells through p21(waf1). Cell Signal 2010; 22:1864-73. [PMID: 20667470 DOI: 10.1016/j.cellsig.2010.07.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 07/08/2010] [Accepted: 07/19/2010] [Indexed: 12/23/2022]
Abstract
The p75 neurotrophin receptor (p75(NTR)) plays a critical role in various neuronal and non-neuronal cell types by regulating cell survival, differentiation and proliferation. To evaluate the influence of p75(NTR) in breast cancer development, we have established and characterized breast cancer cells which stably overexpress p75(NTR). We showed that p75(NTR) overexpression per se promoted cell survival to apoptogens with a concomitant slowdown of cell growth. The pro-survival effect is associated with an increased expression of the inhibitor of apoptosis protein-1 (c-IAP1), a decrease of TRAIL-induced cleavage of PARP, procaspase 9 and procaspase 3, and a decrease of cytochrome C release from the mitochondria. The anti-proliferative effect is due to a cell accumulation in G0/G1, associated with a decrease of Rb phosphorylation and an increase of p21(waf1). Interestingly, inhibition of p21(waf1) with siRNA not only restores proliferation but also abolishes the pro-survival effect of p75(NTR), indicating the key role of p21(waf1) in the biological functions of p75(NTR). Finally, using a SCID mice xenograft model, we showed that p75(NTR) overexpression favors tumor growth and strongly increases tumor resistance to anti-tumoral treatment. Together, our findings suggest that p75(NTR) overexpression in breast tumor cells could favor tumor survival and contribute to tumor resistance to drugs. This provides a rationale to consider p75(NTR) as a potential target for the future design of innovative therapeutic strategies.
Collapse
|
32
|
Gewandter JS, Staversky RJ, O’Reilly MA. Hyperoxia augments ER-stress-induced cell death independent of BiP loss. Free Radic Biol Med 2009; 47:1742-52. [PMID: 19786088 PMCID: PMC2783969 DOI: 10.1016/j.freeradbiomed.2009.09.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 09/05/2009] [Accepted: 09/22/2009] [Indexed: 01/22/2023]
Abstract
Cytotoxic reactive oxygen species are constantly formed as a by-product of aerobic respiration and are thought to contribute to aging and disease. Cells respond to oxidative stress by activating various pathways, whose balance is important for adaptation or induction of cell death. Our lab recently reported that BiP (GRP78), a proposed negative regulator of the unfolded protein response (UPR), declines during hyperoxia, a model of chronic oxidative stress. Here, we investigate whether exposure to hyperoxia, and consequent loss of BiP, activates the UPR or sensitizes cells to ER stress. Evidence is provided that hyperoxia does not activate the three ER stress receptors IRE1, PERK, and ATF6. Although hyperoxia alone did not activate the UPR, it sensitized cells to tunicamycin-induced cell death. Conversely, overexpression of BiP did not block hyperoxia-induced ROS production or increased sensitivity to tunicamycin. These findings demonstrate that hyperoxia and loss of BiP alone are insufficient to activate the UPR. However, hyperoxia can sensitize cells to toxicity from unfolded proteins, implying that chronic ROS, such as that seen throughout aging, could augment the UPR and, moreover, suggesting that the therapeutic use of hyperoxia may be detrimental for lung diseases associated with ER stress.
Collapse
Affiliation(s)
- Jennifer S. Gewandter
- Department of Biochemistry and Biophysics The University of Rochester Rochester, NY 14642
| | | | - Michael A. O’Reilly
- Department of Pediatrics The University of Rochester Rochester, NY 14642
- Address Correspondence to: Michael A. O’Reilly, Ph.D. Department of Pediatrics Box 850 The University of Rochester School of Medicine and Dentistry 601 Elmwood Avenue Rochester NY 14642 Tel: (585) 275-5948 Fax: (585) 756-7780
| |
Collapse
|
33
|
Deslee G, Adair-Kirk TL, Betsuyaku T, Woods JC, Moore CH, Gierada DS, Conradi SH, Atkinson JJ, Toennies HM, Battaile JT, Kobayashi DK, Patterson GA, Holtzman MJ, Pierce RA. Cigarette smoke induces nucleic-acid oxidation in lung fibroblasts. Am J Respir Cell Mol Biol 2009; 43:576-84. [PMID: 20008282 DOI: 10.1165/rcmb.2009-0221oc] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Oxidative stress is widely proposed as a pathogenic mechanism for chronic obstructive pulmonary disease (COPD), but the molecular pathway connecting oxidative damage to tissue destruction remains to be fully defined. We suggest that reactive oxygen species (ROS) oxidatively damage nucleic acids, and this effect requires multiple repair mechanisms, particularly base excision pathway components 8-oxoguanine-DNA glycosylase (OGG1), endonuclease III homologue 1 (NTH1), and single-strand-selective monofunctional uracil-DNA glycosylase 1 (SMUG1), as well as the nucleic acid-binding protein, Y-box binding protein 1 (YB1). This study was therefore designed to define the levels of nucleic-acid oxidation and expression of genes involved in the repair of COPD and in corresponding models of this disease. We found significant oxidation of nucleic acids localized to alveolar lung fibroblasts, increased levels of OGG1 mRNA expression, and decreased concentrations of NTH1, SMUG1, and YB1 mRNA in lung samples from subjects with very severe COPD compared with little or no COPD. Mice exposed to cigarette smoke exhibited a time-dependent accumulation of nucleic-acid oxidation in alveolar fibroblasts, which was associated with an increase in OGG1 and YB1 mRNA concentrations. Similarly, human lung fibroblasts exposed to cigarette smoke extract exhibited ROS-dependent nucleic-acid oxidation. The short interfering RNA (siRNA)-dependent knockdown of OGG1 and YB1 expression increased nucleic-acid oxidation at the basal state and after exposure to cigarette smoke. Together, our results demonstrate ROS-dependent, cigarette smoke-induced nucleic-acid oxidation in alveolar fibroblasts, which may play a role in the pathogenesis of emphysema.
Collapse
Affiliation(s)
- Gaetan Deslee
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Washington University, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Staversky RJ, Vitiello PF, Yee M, Callahan LM, Dean DA, O'Reilly MA. Epithelial ablation of Bcl-XL increases sensitivity to oxygen without disrupting lung development. Am J Respir Cell Mol Biol 2009; 43:376-85. [PMID: 19880821 DOI: 10.1165/rcmb.2009-0165oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Recent studies indicate that the antiapoptotic Bcl-X(L), one of five isoforms expressed by the Bcl-X gene, protects a variety of cell lines exposed to hyperoxia. However, its role in lung development and protection against oxidative stress in vivo is not known. Here, we show Bcl-X(L) is the predominant isoform expressed in the lung, and the only isoform detected in respiratory epithelium. Because loss of Bcl-X(L) is embryonically lethal, Bcl-X(L) was ablated throughout the respiratory epithelium by mating mice with a floxed exon II of the Bcl-X gene with mice expressing Cre under control of the surfactant protein-C promoter. Interestingly, the loss of Bcl-X(L) in respiratory epithelium was perinatally lethal in approximately 50% of the expected offspring. However, some adult mice lacking the gene were obtained. The epithelial-specific ablation of Bcl-X(L) did not disrupt pulmonary function, the expression of epithelial cell-specific markers, or lung development. However, it shifted the lung toward a proapoptotic state, defined by a reduction in antiapoptotic Mcl-1, an increase in proapoptotic Bak, and increased sensitivity of the respiratory epithelium to hyperoxia. Intriguingly, increased 8-oxoguanine lesions seen during hyperoxia were also evident as lungs transitioned to room air at birth, a time when perinatal lethality in some mice lacking Bcl-X(L) was observed. These findings reveal that the epithelial-specific expression of Bcl-X(L) is not required for proper lung development, but functions to protect respiratory epithelial cells against oxygen-induced toxicity, such as during hyperoxia and the lung's first exposure to ambient air.
Collapse
|