• Aucun résultat trouvé

BHV-1 induced oxidative stress contributes to mitochondrial dysfunction in MDBK cells

N/A
N/A
Protected

Academic year: 2021

Partager "BHV-1 induced oxidative stress contributes to mitochondrial dysfunction in MDBK cells"

Copied!
7
0
0

Texte intégral

(1)

HAL Id: hal-01341499

https://hal.archives-ouvertes.fr/hal-01341499

Submitted on 4 Jul 2016

HAL is a multi-disciplinary open access

archive for the deposit and dissemination of sci- entific research documents, whether they are pub- lished or not. The documents may come from teaching and research institutions in France or abroad, or from public or private research centers.

L’archive ouverte pluridisciplinaire HAL, est destinée au dépôt et à la diffusion de documents scientifiques de niveau recherche, publiés ou non, émanant des établissements d’enseignement et de recherche français ou étrangers, des laboratoires publics ou privés.

BHV-1 induced oxidative stress contributes to mitochondrial dysfunction in MDBK cells

Liqian Zhu, Chen Yuan, Dong Zhang, Yan Ma, Xiuyan Ding, Guoqiang Zhu

To cite this version:

Liqian Zhu, Chen Yuan, Dong Zhang, Yan Ma, Xiuyan Ding, et al.. BHV-1 induced oxidative stress

contributes to mitochondrial dysfunction in MDBK cells. Veterinary Research, BioMed Central, 2016,

47 (1), pp.47. �10.1186/s13567-016-0332-2�. �hal-01341499�

(2)

SHORT REPORT

BHV-1 induced oxidative stress

contributes to mitochondrial dysfunction in MDBK cells

Liqian Zhu

1

, Chen Yuan

1

, Dong Zhang

1

, Yan Ma

1

, Xiuyan Ding

1,2

and Guoqiang Zhu

1*

Abstract

The levels of cellular reactive oxygen species (ROS) and ATP as well as the mitochondrial membrane potential (MMP) in response to bovine herpesvirus 1 (BHV-1) infection of MDBK cells were measured, respectively. BHV-1 infection increased ROS production which depended on viral entry, and de novo protein expression and/or DNA replication.

Vice versa, excessive ROS was required for efficient viral replication. Levels of both ATP and MMP were significantly decreased after BHV-1 infection. Interestingly, the loss of MMP was ameliorated by ROS depression. Collectively, ROS dependent mitochondrial damage and ultimately disruption of energy metabolism (ATP depletion) are a potential pathogenic mechanism for BHV-1 infection.

© 2016 Zhu et al. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://

creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/

zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Introduction

Bovine herpesvirus 1 (BHV-1), an enveloped virus belonging to the alphaherpesvirus subfamily, infects cat- tle of all ages and breeds worldwide. Acute infection of BHV-1 results in clinical diseases in the upper respiratory tract, nasal cavity, or ocular cavity [1, 2]. Generally, BHV- 1-induced immune suppression initiates a polymicrobial respiratory tract disease, commonly referred to bovine respiratory disease complex (BRDC), which costs the US cattle industry approximately three billion dollars, annu- ally [2].

Reactive oxidative species (ROS) such as superoxide, hydrogen peroxide (H2O2), peroxynitrite (OONO) and hydroxyl radicals (OH.) have been shown to play impor- tant roles in both physiological and pathophysiological processes. Under physiological conditions, the produc- tion and release of ROS is tightly controlled at specific time and space, which broadly influences cellular pro- cesses such as gene expression, cells proliferation, migra- tion and differentiation [3]. Oxidative stress results from

the imbalance between production of ROS and the pro- tective effect of the antioxidant system responsible for their neutralization and removal [4]. And excessive pro- duction of ROS generally inflicts cell damage through oxidation of macromolecules, such as protein and lipids [5]. Increasing studies suggested that host ROS contrib- uted greatly to the replication of numerous viruses, as well as inflammation-associated tissues damage, such as in the inflammatory diseases caused by influenza virus, Hepatitis C virus and Dengue virus [6–10]. Thus, sup- pression of ROS appears to be a potential therapeutic approach primarily through reduction of both viral bur- den and inflammatory response.

It has been demonstrated that BHV-1 infection of MDBK cells induced apoptotic responses through diverse mechanisms including the mitochondria-mediate path- way [11, 12]. And mitochondrial damage is a potential indicator of cell apoptotic response. To our knowledge what happed to the mitochondria in BHV-1 infected MDBK cells is unknown.

In this study, we demonstrated that high level of cel- lular ROS was dramatically induced due to BHV-1 virus entry, de novo protein expression and/or DNA replica- tion. The increased ROS was a crucial cellular signaling contributed to viral replication and mitochondrial dam- age, which consequently interfered with cellular ATP

Open Access

*Correspondence: yzgqzhu@yzu.edu.cn

1 College of Veterinary Medicine and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 Wenhui East Road, Yangzhou 225009, China

Full list of author information is available at the end of the article

(3)

Page 2 of 6 Zhu et al. Vet Res (2016) 47:47

production. Our results suggested that the enhanced ROS production by BHV-1 infection is a double-edged sword, which not only promoted the viral infection, but also brought damage to the cells due to the injury to mitochondrial as well as ATP depletion. This could be a potential mechanism for the pathogenicity of BHV-1 infection induced tissue damage.

Materials, methods and results

To evaluate whether BHV-1 infection triggered intracel- lular ROS production, MDBK cells (kindly provided by Dr Leonard J. Bello, University of Pennsylvania) were infected with BHV-1(Colorado 1 strain, kindly provided by Dr Leonard J. Bello) (MOI = 1) at 37 °C for 1, 5 and 12 h.

We selected these time points because BHV-1 replication starts within 2 h of infection in cattle, with cell surface anti- gen expression within 3–4 h after infection and viral release and spread starting 8 h after infection [13]. For the control, the cells were mock infected with supernatant of cell cul- tures that were made by frozen-thawing and subsequent centrifugation using a procedure similar to the generation of virus stocks. The level of intracellular ROS was deter- mined using ROS fluorescence indicator 2′,7′–dichlorodi- hydrofluorescein diacetate (H2DCFDA) (Sigma-Aldrich, St. Louis, MO, USA) which can be converted to fluores- cent compound dichlorofluorescein (DCF). The images were acquired under a fluorescence microscope (Olympus BX-51, Olympus, Tokyo, Japan), and fluorescence intensity was quantified using Image-Pro Plus 6 software. Compared to the control, BHV-1 infection of MDBK cells indeed sig- nificantly increased the production of intracellular ROS at all the detected time points (Figure 1A). Since cellular ROS was significantly increased by the viral infection at 1 h post- infection (pi), ultraviolet (UV)-inactivated viral particles, a replication deficient virus that could enter cells but could not finish subsequent replication steps, was employed to elucidate whether viral entry process increase cellular ROS.

The (UV)-inactivated viral particles showed similar capacity to that of live virus to induce ROS at 1 h pi, but it declined to a level near the mock infected control at 5 and 12 h pi (Figure 1A).

Acyclovir (ACY) is a known inhibitor to suppress her- pesviruses DNA synthesis [14]. The relation between BHV- 1-induced ROS production and viral DNA synthesis was investigated through treatment of MDBK cells with ACY.

MDBK cells were pretreated with ACY (Sigma-Aldrich, St. Louis, MO, USA) at concentrations ranged from 100 to 5 mM for 1 h, and then infected with BHV-1 at MOI of 10 along with the inhibitor. At 4 h pi the cellular ROS was

1 h pi 5 h pi 12 h pi 0

1 2

3 Control

BHV-1 UV-BHV-1

**

*

* ns

ns

ROS fluorescence (fold change vs. control)

Control BHV-1 0.0

0.5 1.0 1.5 2.0 2.5

* *

* ns

ROS fluorescence (fold change vs. control)

Control BHV-1 0.0

0.5 1.0 1.5 2.0 2.5

*

*

* ns

ROS fluorescence (fold change vs. control)

B A

C

Figure 1 Intracellular ROS was increased following BHV-1 infection. A MDBK cells were infected with UV-inactivated or untreated BHV-1 at MOI of 10. At indicated time point, H2DCFDA (5 μM, 30 min) was loaded for visualization of ROS with fluorescence microscopy. B and C MDBK cells pretreated with inhibitor ACY (B) or PAA (C) at indicated concentrations were infected with BHV-1 (MOI = 10) in the presence of corresponding inhibitor. At 4 h pi intracellular ROS were detected with H2DCFDA. Quantification of fluorescence intensity was performed using Image-pro Plus 6. The measured fluorescence intensity were expressed as a relative fold change vs. that in the control cells. Data represent three independ- ent experiments with triplicates performed at each time. Statistical analyses were performed using Student’s t test (Asterisk indicates P < 0.05 vs. control).

(4)

detected with H2DCFDA. As a result, cellular ROS induced by viral infection was significantly depleted by ACY with a dose-dependent manner (Figure 1B). Phosphonoacetic acid (PAA), is a more specific inhibitor targeting the viral DNA polymerase [15]. Treatment of MDBK cells with chemi- cal PAA  (Sigma-Aldrich) at concentrations ranged from 500 to 50 μM with the same manner as that with ACY led to a reduction of viral induced ROS with a dose-depend- ent manner (Figure 1C). It is correlated with the result of MDBK cells treated with ACY. Thus, de novo viral protein production and/or DNA replication seems to be correlated to the elevated ROS levels, which was further confirmed by the analysis with UV-inactivated viral particles.

NADPH oxidases (NOXs) are major sources of ROS pro- duction, which are implicated in multiple viral infections, such as in influenza virus infection [8, 16]. Here, the widely used NOX inhibitor diphenylene iodonium (DPI) and ROS scavenger N-Acetyl-l-cysteine (NAC) were employed to assess the effect of cellular ROS on BHV-1 replication in MDBK cells. These cells were pretreated with DPI (Sigma- Aldrich) or NAC (Sigma-Aldrich) at indicated concentra- tions for 1 h, and then infected with BHV-1. At 4 h pi and 24 h pi cellular ROS and viral titer were detected, respec- tively. The treatment of MDBK cells with both DPI and NAC at indicated concentrations resulted in significant reduc- tion of BHV-1-induced cellular ROS as well as the progeny virus yield with a dose-dependent manner, respectively (Figures 2A–D). The compound at the tested concentrations didn’t show apparent cytotoxicity to MDBK cells as deter- mined by MTT (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide, provided by Sigma-Aldrich) assay performed as we previously described [17] (Figure 2E). It ruled out the possibility that reduced virus titer was caused by strong cellular toxicity of chemicals. Obviously, the increased cellular ROS was required for BHV-1 replication.

To assess if BHV-1 infection caused mitochondrial dys- function, MDBK cells of confluent in 96-well plates were mock infected with DMEM medium containing cell lysis or infected with BHV-1 (MOI = 0.01). Carbonyl cyanide- m-chlorophenyl hydrazone (CCCP) is a known chemical causing rapid mitochondrial depolarization  [18]. Here, CCCP (Beyotime Biotechnology, Jiangsu, China) was introduced to treat the cells as a positive control. At 12, 24, 36 and 48 h pi, the cells were washed twice with PBS, incubated with 3 μg/mL of 5,5′,6,6′-Tetrachloro-1,1′,3,3′- tetraethylbenzimidazolyl-carbocyanine iodide (JC-1) (Beyotime Biotechnology, Jiangsu, China) for 20  min at 37 °C. JC-1 dye is a widely used indicator of mitochon- drial polarization [19, 20]. In healthy cells with higher mitochondrial potential, JC-1 forms a red-fluorescent aggregate (dimer). In unhealthy cells with lower mito- chondrial membrane potential, JC-1 remains in the cyto- plasm as green—fluorescent monomers resulting in a

decrease in the red/green ratio. The decrease in the ratio of red/green fluorescence at 48  h pi following BHV-1 infection indicated the loss of mitochondrial membrane potential (MMP), which showed a level similar to the positive control treated with 1 μM of CCCP (Figure 3A).

In view of that overproduction of cellular ROS may exhibit toxic effect on the mitochondria, MDBK cells were treated with both NAC and Glutathione (GSH, provided by Beyo- time Biotechnology, Jiangsu, China), a main intracellular non-enzymatic antioxidant exerting an efficient buffering role against ROS during the virus infection, to characterize the relation between ROS and the loss of MMP. As a result, both NAC (at a concentration of 5 mM) and GSH (at con- centrations of 5 μM and 10 μM) significantly ameliorated the drop of MMP induced by BHV-1 infection at 48 h pi (Figure 3B). It implied that the virus triggered overproduc- tion of ROS was partially associated with the loss of MMP.

Because ATP is mainly produced in the mitochondria which is associated with energy metabolism, the loss of ATP content is considered to be a powerful indicator of mitochondrial dysfunction. Confluent MDBK cells in 6-well plates were mock infected with DMEM contain- ing supernatant of cell culture or infected with BHV-1 (MOI  =  0.01). At 12, 24 and 48  h pi, ATP content was determined with a commercial luminescence kit (Beyo- time Biotechnology, Jiangsu, China), the intensity was read with multifunctional Microplate Reader (SpectraMax M2, MDC) and normalized to the respective protein con- centrations in the cell lysis detected with Bradford Pro- tein Assay Kit (Beyotime Biotechnology, Jiangsu, China).

Interestingly, comparing to the control, ATP content was significantly decreased at 48 h pi in MDBK cells (Figure 4), which correlated well with the reduction of MMP by BHV-1 infection (Figure 3A). It further confirmed that the virus infection led to mitochondrial dysfunction.

Discussion

Overproduced cellular ROS or oxidative stress is a criti- cal factor that influences disease outcome, in the case of multiple viral infection such as by infection of influenza virus and Hepatitis C virus [6, 8, 21]. In this study, we found that BHV-1 infection of MDBK cells dramatically increased the production of cellular ROS, which is essen- tial for virus replication (Figures 1 and 2).

BHV-1 together with Herpes simplex virus (HSV) and HSV-2 all belongs to the Alphaherpesvirinae subfamily, and shares a number of biological properties. The inter- plays between ROS and HSV-1 or HSV-2 in various cell cultures have been extensively studied [22–24]. Based on both our studies and studies from others [23–25], we inferred that cellular ROS is broadly regulated by alpha herpesviruses, but diverse manners were presented, which are cell type and virus specific, e.g., HSV-2 induced

(5)

Page 4 of 6 Zhu et al. Vet Res (2016) 47:47

Mock BHV-1 BHV-1 + 0.1 mM NAC

BHV-1 + 5 mM NAC BHV-1 + 10 mM NAC 0

1 2 3 4

*

* ns *

ROS fluorescence (Fold change vs. mockl)

Mock BHV-1 0

1 2 3

*

* ns ns

ROS fluoresence (Fold change vs. control)

Mock

0.1 mM NAC

5 mM NAC

10 mM NAC 0

2 4 6

8 *

ns *

Virus titer log 10

Mock 0 1 2 3 4 5

* ns

ns

Virus titer log 10

Mock

10 mM NAC 0

20 40 60 80 100 120

% of viable cells vs. control

A B

C D

E

Figure 2 The reduction of ROS levels by both ROS scavenger NAC and NOXs inhibitor DPI inhibited BHV-1 replication in MDBK cells.

(A and B) MDBK cells subjected to a pretreatment with DMSO control or chemicals NAC (A) or DPI (B) for 1 h, were mock infected with supernatant of cell culture or infected with BHV-1 along with chemical treatment. At 4 h pi cellular ROS were detected using H2DCFDA (5 μM, 30 min), and the quantification of fluorescence intensity was performed using Image-pro Plus 6. (C and D) MDBK cells infected with BHV-1 (MOI = 1) were treated with chemicals including NAC (C) or DPI (D) during viral infection plus a pretreatment for 1 h. At 24 h pi the virus yields were determined with TCID50 method. (E) Cell viability of MDBK cells exposed to each inhibitor as designated concentrations was detected with MTT assay. Values repre- sent three independent experiments. Statistical analyses of significant difference vs. mock were performed using Student’s t test (Asterisk indicates P < 0.05 vs. mock).

(6)

ROS production in RAW246.7 cells could be uniquely detected at 1 h pi [25], HSV-1 infection of murine micro- glial cells and neural cells increased ROS levels at time durations of 24–72 and 1–24 h pi, respectively [23, 24].

Increased ROS levels in MDBK cells was detected at 1–12 h pi during BHV-1 infection (Figure 1), and also it appeared at 24 h pi (data not shown). It is highly possible that ROS have diverse effect on viral replication or even on viral pathogenesis.

ROS mainly originated from Nox family of NADPH oxidases and mitochondria. Mitochondria are also susceptible to insult from ROS, since both mitochon- drial proteins and mitochondrial DNA are vulnerable to ROS [26]. It is known that mitochondria is a poten- tial target of oxidative stress for environmental toxi- cants stimulus like the fine particulate matter (PM2.5) [27]. It is reasonable that mitochondria is a potential target by some viruses that are able to induce exces- sive ROS. It has been documented that HSV infection results in the loss of MMP and decreases the levels of cellular ATP at the late stage of infection [28], whereas the role of ROS in this adverse effect is not yet eluci- dated. In this study, we found that BHV-1 infection of MDBK cells rendered mitochondria dysfunction as demonstrated by the mitochondrial depolarization and reduced ATP levels at late stage of infection. The loss of MMP was partially reversed by antioxidants of GSH and ROS scavenger NAC, which indicated that the overexpressed ROS partially accounts for the mito- chondrial dysfunction.

Currently, the pathogenic mechanism of ROS in viral disease mainly reside in the ROS—mediated promotion of viral replication and inflammatory response. We sug- gested that BHV-1 induced ROS/mitochondrial damage and ultimately interfered with energy metabolism (as demonstrated by ATP depletion) is a novel mechanism by which BHV-1 induced cell injury. In addition to ROS mediated inflammatory response, this is also a potential pathogenic mechanism employed by alpha herpesvirus infection, such as in BHV-1 infection.

Competing interests

The authors declare that they have no competing interests.

Author details

1 College of Veterinary Medicine and Jiangsu Co-innovation Center for Pre- vention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, 48 Wenhui East Road, Yangzhou 225009, China. 2 The Test Center of Yangzhou University, 48 Wenhui East Road, Yangzhou 225009, China.

Authors’ contributions

LZ participated in design of the study, analyzed the data and prepared the manuscript. CY, DZ, YM and XD carried out the experiments, GZ revised the manuscript and coordinated the research. All authors read and approved the final manuscript.

12 h 24 h 36 h 48 h

0 50 100 150

Mock BHV-1 Positive control

ns* * * *

ns ns *

MMP (% of control)

Mock BHV-1

BHV-1+1 mM NACBHV-1+5 mM NA C 0

50 100 150

*

* * ns * MMP (% of control)

A

B

Figure 3 Effect of BHV-1 on mitochondrial membrane poten- tial in MDBK cells. (A) MDBK cells of confluent in 96-well plates were infected with BHV-1 at MOI of 0.01 for indicated time length, and then MMP was detected with JC-1 dye. The treatment of cells with CCCP for 20 min was used as a positive control. (B) MDBK cells of confluent in 96-well plates were treated with NAC and GSH at indi- cated concentrations, before and during virus infection with BHV-1 (MOI = 0.01) for 48 h. The MMP was detected with JC-1 dye. Data represent three independent experiments. Statistical analyses were performed using Student’s t test (Asterisk indicates P < 0.05 vs. mock).

12 h pi 24 h pi 48 h pi

0 50 100

150 Control BHV-1

ns ns *

ATP content (% of control)

Figure 4 Effect of BHV-1 on ATP content during the infec- tion of MDBK cells. MDBK cells of confluent in 6-well plates were infected with BHV-1 at MOI of 0.01 for indicated time length. The content of ATP and protein concentration was determined with ATP Assay Kit and Bradford Protein Assay Kit, respectively. The ATP content was normalized to the protein concentrations and expressed as a

% of mock treated control cells. Data represent three independent experiments. Statistical analyses were performed using Student’s t test (Asterisk indicates P < 0.05 vs. control).

(7)

Page 6 of 6 Zhu et al. Vet Res (2016) 47:47

• We accept pre-submission inquiries

• Our selector tool helps you to find the most relevant journal

• We provide round the clock customer support

• Convenient online submission

• Thorough peer review

• Inclusion in PubMed and all major indexing services

• Maximum visibility for your research Submit your manuscript at

www.biomedcentral.com/submit

Submit your next manuscript to BioMed Central and we will help you at every step:

Acknowledgements

The authors are grateful to Dr Leonard J. Bello, University of Pennsylvania, for providing the MDBK cells and the Colorado 1 strain of BHV-1. This research was supported by Chinese National Science Foundation Grant (No. 31472172), and partially by the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD), and Natural Science Foundation from Chongqing (cstc2014jcyjA1628), Grants No. 14KJA230001 and No. BE2014358 from the Jiangsu province, and 948 program from Ministry of Agriculture of the People’s Republic of China (Grant No. 2011-G24).

Received: 22 November 2015 Accepted: 4 March 2016

References

1. Jones C (2009) Regulation of innate immune responses by bovine her- pesvirus 1 and infected cell protein 0 (bICP0). Viruses 1:255–275 2. Jones C, Chowdhury S (2007) A review of the biology of bovine herpesvi-

rus type 1 (BHV-1), its role as a cofactor in the bovine respiratory disease complex and development of improved vaccines. Animal Health Res Rev 8:187–205

3. Chen F, Yu Y, Haigh S, Johnson J, Lucas R, Stepp DW, Fulton DJ (2014) Regulation of NADPH oxidase 5 by protein kinase C isoforms. PLoS One 9:e88405

4. Walczak-Jedrzejowska R, Wolski JK, Slowikowska-Hilczer J (2013) The role of oxidative stress and antioxidants in male fertility. Cent European J Urol 66:60–67

5. Hussain SP, Hofseth LJ, Harris CC (2003) Radical causes of cancer. Nat Rev Cancer 3:276–285

6. Gonzalez-Gallego J, Garcia-Mediavilla MV, Sanchez-Campos S (2011) Hepatitis C virus, oxidative stress and steatosis: current status and per- spectives. Cur Mol Med 11:373–390

7. Ye S, Lowther S, Stambas J (2015) Inhibition of reactive oxygen species production ameliorates inflammation induced by influenza A viruses via upregulation of SOCS1 and SOCS3. J Virol 89:2672–2683

8. Amatore D, Sgarbanti R, Aquilano K, Baldelli S, Limongi D, Civitelli L, Nen- cioni L, Garaci E, Ciriolo MR, Palamara AT (2015) Influenza virus replication in lung epithelial cells depends on redox-sensitive pathways activated by NOX4-derived ROS. Cell Microbiol 17:131–145

9. Salhan D, Husain M, Subrati A, Goyal R, Singh T, Rai P, Malhotra A, Singhal PC (2012) HIV-induced kidney cell injury: role of ROS-induced downregu- lated vitamin D receptor. Am J Physiol Renal Physiol 303:F503–F514 10. Olagnier D, Peri S, Steel C, van Montfoort N, Chiang C, Beljanski V, Slifker

M, He Z, Nichols CN, Lin R, Balachandran S, Hiscott J (2014) Cellular oxidative stress response controls the antiviral and apoptotic programs in dengue virus-infected dendritic cells. PLoS Pathog 10:e1004566 11. Devireddy LR, Jones CJ (1999) Activation of caspases and p53 by bovine

herpesvirus 1 infection results in programmed cell death and efficient virus release. J Virol 73:3778–3788

12. Xu X, Zhang K, Huang Y, Ding L, Chen G, Zhang H, Tong D (2012) Bovine herpes virus type 1 induces apoptosis through Fas-dependent and mitochondria-controlled manner in Madin-Darby bovine kidney cells.

Virol J 9:202

13. Babiuk LA, van Drunen Littel-van den Hurk S, Tikoo SK (1996) Immunol- ogy of bovine herpesvirus 1 infection. Vet Microbiol 53:31–42 14. Wuest T, Zheng M, Efstathiou S, Halford WP, Carr DJ (2011) The herpes

simplex virus-1 transactivator infected cell protein-4 drives VEGF-A dependent neovascularization. PLoS Pathog 7:e1002278

15. Becker Y, Asher Y, Cohen Y, Weinberg-Zahlering E, Shlomai J (1977) Phosphonoacetic acid-resistant mutants of herpes simplex virus: effect of phosphonoacetic acid on virus replication and in vitro deoxyribonu- cleic acid synthesis in isolated nuclei. Antimicrob Agents Chemother 11:919–922

16. Montezano AC, Touyz RM (2012) Reactive oxygen species and endothelial function–role of nitric oxide synthase uncoupling and Nox family nicoti- namide adenine dinucleotide phosphate oxidases. Basic Clin Pharmacol Toxicol 110:87–94

17. Zhu L, Ding X, Zhu X, Meng S, Wang J, Zhou H, Duan Q, Tao J, Schifferli DM, Zhu G (2011) Biphasic activation of PI3 K/Akt and MAPK/Erk1/2 signaling pathways in bovine herpesvirus type 1 infection of MDBK cells.

Vet Res 42:57

18. Nieminen AL, Saylor AK, Herman B, Lemasters JJ (1994) ATP depletion rather than mitochondrial depolarization mediates hepatocyte killing after metabolic inhibition. Am J Physiol 267:C67–C74

19. Guo X, Chen S, Zhang Z, Dobrovolsky VN, Dial SL, Guo L, Mei N (2015) Reactive oxygen species and c-Jun N-terminal kinases contribute to TEMPO-induced apoptosis in L5178Y cells. Chem Biol Interact 235:27–36 20. Yoshizumi T, Ichinohe T, Sasaki O, Otera H, Kawabata S, Mihara K, Koshiba T (2014) Influenza A virus protein PB1-F2 translocates into mitochondria via Tom40 channels and impairs innate immunity. Nat Commun 5:4713 21. Anand SK, Tikoo SK (2013) Viruses as modulators of mitochondrial func-

tions. Adv Virol 2013:738794

22. Schachtele SJ, Hu S, Lokensgard JR (2012) Modulation of experimental herpes encephalitis-associated neurotoxicity through sulforaphane treat- ment. PLoS One 7:e36216

23. Hu S, Sheng WS, Schachtele SJ, Lokensgard JR (2011) Reactive oxygen species drive herpes simplex virus (HSV)-1-induced proinflammatory cytokine production by murine microglia. J Neuroinflammation 8:123 24. Kavouras JH, Prandovszky E, Valyi-Nagy K, Kovacs SK, Tiwari V, Kovacs

M, Shukla D, Valyi-Nagy T (2007) Herpes simplex virus type 1 infection induces oxidative stress and the release of bioactive lipid peroxidation by-products in mouse P19 N neural cell cultures. J Neurovirol 13:416–425 25. Gonzalez-Dosal R, Horan KA, Rahbek SH, Ichijo H, Chen ZJ, Mieyal JJ,

Hartmann R, Paludan SR (2011) HSV infection induces production of ROS, which potentiate signaling from pattern recognition receptors: role for S-glutathionylation of TRAF3 and 6. PLoS Pathog 7:e1002250 26. Shaughnessy DT, McAllister K, Worth L, Haugen AC, Meyer JN, Domann

FE, Van Houten B, Mostoslavsky R, Bultman SJ, Baccarelli AA, Begley TJ, Sobol RW, Hirschey MD, Ideker T, Santos JH, Copeland WC, Tice RR, Balshaw DM, Tyson FL (2014) Mitochondria, energetics, epigenetics, and cellular responses to stress. Environ Health Perspect 122:1271–1278 27. Li R, Kou X, Geng H, Xie J, Tian J, Cai Z, Dong C (2015) Mitochondrial

damage: an important mechanism of ambient PM2.5 exposure-induced acute heart injury in rats. J Hazard Mater 287:392–401

28. Murata T, Goshima F, Daikoku T, Inagaki-Ohara K, Takakuwa H, Kato K, Nishiyama Y (2000) Mitochondrial distribution and function in herpes simplex virus-infected cells. J Gen Virol 81:401–406

Références

Documents relatifs

 ISSEA, Institut Sous-régional de Statistique et d'Économie Appliquée de Yaoundé  ENSAE, École Nationale de la Statistique et de l'Analyse Économique de Dakar  ENEAM,

Mitochondrial dysfunction results from oxidative stress in the skeletal muscle of diet-induced insulin- resistant mice.... Intraperitoneal glucose (A) and insulin (B) tolerance tests

In this study, we observed that the expression of Bfl-1, an antiapoptotic protein of the Bcl-2 family, is restricted to HTLV-1-infected T-cell lines and to T-cells expressing both

In summary, for the first time we elucidated that in BHV-1 infected MDBK cells, all three major MAPK pathways are activated in response to viral infection, but JNK signaling

To address this problem, in this study, we tested the immunomodulatory activity of the novel sensitive mAbs against bovine PD-L1 and LAG-3 established in recent studies [ 20 , 21

Pour conclure, une façon efficace d’établir les fondations d’une éducation de qualité en Afrique consiste à utiliser un support d’instruction familier pendant une période d’au

sorbitol, pBK-CMV-UL14 plasmid transfected MDBK cells (UL14 positive), empty vector 22.. transfected MDBK cells, and non-transfected/not-treated MDBK cells (UL14 negative) were

B[a]P/ethanol coexposure reduced mitochondrial respiratory chain activity, mitochondrial respiration, and mitochondrial DNA levels and induced ROS overproduction in steatotic