Your browser doesn't support javascript.
loading
Mast cells regulate CD4+ T-cell differentiation in the absence of antigen presentation.
Rodriguez Cetina Biefer, Hector; Heinbokel, Timm; Uehara, Hirofumi; Camacho, Virginia; Minami, Koichiro; Nian, Yeqi; Koduru, Suresh; El Fatimy, Rachid; Ghiran, Ionita; Trachtenberg, Alexander J; de la Fuente, Miguel A; Azuma, Haruhito; Akbari, Omid; Tullius, Stefan G; Vasudevan, Anju; Elkhal, Abdallah.
Afiliación
  • Rodriguez Cetina Biefer H; Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Clinic for Cardiovascular Surgery, University Hospital Zurich, Zurich, Switzerland.
  • Heinbokel T; Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Department of Nephrology, Charite Universitaetsmedizin Berlin, Berlin, Germany.
  • Uehara H; Department of Urology, Osaka Medical College, Osaka, Japan.
  • Camacho V; Flow Cytometry Core Facility, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Boston, Mass.
  • Minami K; Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass.
  • Nian Y; Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass.
  • Koduru S; School of Medical Sciences, University of Hyderabad, Hyderabad, India.
  • El Fatimy R; Department of Neurology, Center for Neurologic Diseases, Initiative for RNA Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass.
  • Ghiran I; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Boston, Mass.
  • Trachtenberg AJ; StART Families, Boston, Mass.
  • de la Fuente MA; Instituto de Biología y Genética Molecular, University of Valladolid, Valladolid, Spain.
  • Azuma H; Department of Urology, Osaka Medical College, Osaka, Japan.
  • Akbari O; Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif.
  • Tullius SG; Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass.
  • Vasudevan A; Angiogenesis and Brain Development Laboratory, Division of Basic Neuroscience, McLean Hospital, Harvard Medical School, Belmont, Mass.
  • Elkhal A; Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass. Electronic address: aelkhal@partners.org.
J Allergy Clin Immunol ; 142(6): 1894-1908.e7, 2018 12.
Article en En | MEDLINE | ID: mdl-29470999
ABSTRACT

BACKGROUND:

Given their unique capacity for antigen uptake, processing, and presentation, antigen-presenting cells (APCs) are critical for initiating and regulating innate and adaptive immune responses. We have previously shown the role of nicotinamide adenine dinucleotide (NAD+) in T-cell differentiation independently of the cytokine milieu, whereas the precise mechanisms remained unknown.

OBJECTIVE:

The objective of this study is to further dissect the mechanism of actions of NAD+ and determine the effect of APCs on NAD+-mediated T-cell activation.

METHODS:

Isolated dendritic cells and bone marrow-derived mast cells (MCs) were used to characterize the mechanisms of action of NAD+ on CD4+ T-cell fate in vitro. Furthermore, NAD+-mediated CD4+ T-cell differentiation was investigated in vivo by using wild-type C57BL/6, MC-/-, MHC class II-/-, Wiskott-Aldrich syndrome protein (WASP)-/-, 5C.C7 recombination-activating gene 2 (Rag2)-/-, and CD11b-DTR transgenic mice. Finally, we tested the physiologic effect of NAD+ on the systemic immune response in the context of Listeria monocytogenes infection.

RESULTS:

Our in vivo and in vitro findings indicate that after NAD+ administration, MCs exclusively promote CD4+ T-cell differentiation, both in the absence of antigen and independently of major APCs. Moreover, we found that MCs mediated CD4+ T-cell differentiation independently of MHC II and T-cell receptor signaling machinery. More importantly, although treatment with NAD+ resulted in decreased MHC II expression on CD11c+ cells, MC-mediated CD4+ T-cell differentiation rendered mice resistant to administration of lethal doses of L monocytogenes.

CONCLUSIONS:

Collectively, our study unravels a novel cellular and molecular pathway that regulates innate and adaptive immunity through MCs exclusively and underscores the therapeutic potential of NAD+ in the context of primary immunodeficiencies and antimicrobial resistance.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Linfocitos T CD4-Positivos / Mastocitos / NAD Límite: Adult / Animals / Humans Idioma: En Revista: J Allergy Clin Immunol Año: 2018 Tipo del documento: Article País de afiliación: Suiza

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Linfocitos T CD4-Positivos / Mastocitos / NAD Límite: Adult / Animals / Humans Idioma: En Revista: J Allergy Clin Immunol Año: 2018 Tipo del documento: Article País de afiliación: Suiza