4.6 Article

Generation of NO by Bystander Human CD8 T Cells Augments Allogeneic Responses by Inhibiting Cytokine Deprivation-Induced Cell Death

Journal

AMERICAN JOURNAL OF TRANSPLANTATION
Volume 9, Issue 10, Pages 2281-2291

Publisher

WILEY
DOI: 10.1111/j.1600-6143.2009.02771.x

Keywords

Cell death; endothelial cells; nitric oxide; T cell

Funding

  1. National Institutes of Health [HL070295]
  2. Canadian Institutes of Health Research
  3. International Society for Heart and Lung Transplantation

Ask authors/readers for more resources

Nitric oxide (NO), generated by inducible NO synthase (iNOS) in bystander human CD8 T cells, augments the accumulation of allogeneically activated human CD8 T cells in vitro and in vivo. Here, we report that iNOS-derived NO does not affect T-cell proliferation but rather inhibits cell death of activated human CD8 T cells after activation by allogeneic endothelial cells in culture. Exogenous NO did not affect activation-induced cell death of human CD8 T cells but specifically reduced death of activated T cells due to cytokine deprivation. NO-mediated inhibition of T-cell death did not involve cGMP signaling, and NO did not affect the expression of Bcl-2-related proteins known to regulate cytokine deprivation-induced cell death. However, NO inhibited the activity of caspases activated as a consequence of cytokine deprivation in activated T cells. This protective effect correlated with S-nitrosylation of caspases and was phenocopied by z-VAD.fmk and z-LEHD.fmk, pharmacological inhibitors of caspases. In summary, our findings indicate that NO augments the accumulation of activated human T cells principally by inhibiting cytokine deprivation-induced cell death through S-nitrosylation of caspases.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

Article Multidisciplinary Sciences

Mouse Model of Alloimmune-induced Vascular Rejection and Transplant Arteriosclerosis

Winnie Enns, Anna von Rossum, Jonathan Choy

JOVE-JOURNAL OF VISUALIZED EXPERIMENTS (2015)

Review Immunology

Immune-mediated vascular injury and dysfunction in transplant arteriosclerosis

Anna von Rossum, Ismail Laher, Jonathan C. Choy

FRONTIERS IN IMMUNOLOGY (2015)

Article Immunology

Graft-Derived IL-6 Amplifies Proliferation and Survival of Effector T Cells That Drive Alloimmune-Mediated Vascular Rejection

Anna von Rossum, Kevin Rey, Winnie Enns, Sukhbir Manku, Rajan Cheema, Grace E. MacEwan, Jonathan C. Choy

TRANSPLANTATION (2016)

Article Cardiac & Cardiovascular Systems

IL-6 expression is correlated with increased T-cell proliferation and survival in the arterial wall in giant cell arteritis

Sukhbir Manku, Wendy Wong, Zongshu Luo, Michael A. Seidman, Zainab Alabdurubalnabi, Kevin Rey, Winnie Enns, J. Antonio Avina-Zubieta, Kamran Shojania, Jonathan C. Choy

CARDIOVASCULAR PATHOLOGY (2018)

Article Immunology

Disruption of the Gut Microbiota With Antibiotics Exacerbates Acute Vascular Rejection

Kevin Rey, Sukhbir Manku, Winnie Enns, Thea Van Rossum, Kevin Bushell, Ryan D. Morin, Fiona S. L. Brinkman, Jonathan C. Choy

TRANSPLANTATION (2018)

Article Microbiology

The Aspergillus fumigatus Sialidase (Kdnase) Contributes to Cell Wall Integrity and Virulence in Amphotericin B-Treated Mice

Jason R. Nesbitt, Elizabeth Y. Steves, Cole R. Schonhofer, Alissa Cait, Sukhbir S. Manku, Juliana H. F. Yeung, Andrew J. Bennet, Kelly M. McNagny, Jonathan C. Choy, Michael R. Hughes, Margo M. Moore

FRONTIERS IN MICROBIOLOGY (2018)

Article Hematology

CD155 on Human Vascular Endothelial Cells Attenuates the Acquisition of Effector Functions in CD8 T Cells

Nichole K. Escalante, Anna von Rossum, Martin Lee, Jonathan C. Choy

ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY (2011)

Article Hematology

Bim Regulates Alloimmune-Mediated Vascular Injury Through Effects on T-Cell Activation and Death

Anna von Rossum, Winnie Enns, Yu P. Shi, Grace E. MacEwan, Mehrnoush Malekesmaeli, Ryan Brinkman, Jonathan C. Choy

ARTERIOSCLEROSIS THROMBOSIS AND VASCULAR BIOLOGY (2014)

Review Biochemistry & Molecular Biology

Granzymes and perforin in solid organ transplant rejection

J. C. Choy

CELL DEATH AND DIFFERENTIATION (2010)

Article Biochemistry & Molecular Biology

Inflammatory Cytokines Determine the Susceptibility of Human CD8 T Cells to Fas-mediated Activation-induced Cell Death through Modulation of FasL and c-FLIPS Expression

Anna von Rossum, Randall Krall, Nichole K. Escalante, Jonathan C. Choy

JOURNAL OF BIOLOGICAL CHEMISTRY (2011)

Article Biochemistry & Molecular Biology

Positive Feedback Regulation of Human Inducible Nitric-oxide Synthase Expression by Ras Protein S-Nitrosylation

Martin Lee, Jonathan C. Choy

JOURNAL OF BIOLOGICAL CHEMISTRY (2013)

Article Immunology

IFN-γ Primes Intact Human Coronary Arteries and Cultured Coronary Smooth Muscle Cells to Double-Stranded RNA- and Self-RNA-Induced Inflammatory Responses by Upregulating TLR3 and Melanoma Differentiation-Associated Gene 5

Usman Ahmad, Rahmat Ali, Amir H. Lebastchi, Lingfeng Qin, Sheng-fu L. Lo, Alexander O. Yakimov, Salman F. Khan, Jonathan C. Choy, Arnar Geirsson, Jordan S. Pober, George Tellides

JOURNAL OF IMMUNOLOGY (2010)

Article Medical Laboratory Technology

Immunologic Effects of the Microbiota in Organ Transplantation

Kevin Rey, Jonathan C. Choy

CLINICS IN LABORATORY MEDICINE (2019)

Article Biochemistry & Molecular Biology

Expression of human inducible nitric oxide synthase in response to cytokines is regulated by hypoxia-inducible factor-1

Martin Lee, Christine Wang, Steven W. Jin, Mark P. Labrecque, Timothy V. Beischlag, Mark A. Brockman, Jonathan C. Choy

FREE RADICAL BIOLOGY AND MEDICINE (2019)

No Data Available