Upregulation of CD47 is a host checkpoint response to pathogen recognition

Autor: Mark M. Davis, Lamin B. Cham, Thai Nguyen, Ronald J. Messer, Benjamin Fram, Kim J. Hasenkrug, Katrin D. Mayer-Barber, Cesar J. Lopez Angel, Karin E. Peterson, Denise M. Monack, Aijaz Ahmed, Maxim Markovic, Michal Caspi Tal, Lara Myers, Laughing Bear Torrez Dulgeroff, Debashis Sahoo, Irving L. Weissman, Jens Kortmann, Karl S. Lang, Tom Adomati, Jayakumar Rajadas, Jeffrey S. Glenn, Raja Kalluru, Clayton W. Winkler, Ed Pham, Ehydel Castro, Ying Ying Yiu, Eric Gars, Daniel M. Corey, Sarah Danielle Galloway, Niaz Banaei, Tyson A. Woods, Dhananjay Wagh, Paige Hansen, Michaela Hasenkrug, Aaron B. Carmody, Andrea C. Bohrer
Přispěvatelé: Vance, Russell
Jazyk: angličtina
Rok vydání: 2020
Předmět:
Male
animal diseases
medicine.medical_treatment
Medizin
Adaptive Immunity
Inbred C57BL
Mice
0302 clinical medicine
Receptors
Lymphocytic choriomeningitis virus
Innate
2.1 Biological and endogenous factors
host response
Aetiology
CD47
Lung
innate immunity
Mice
Knockout

0303 health sciences
Tumor
Pattern recognition receptor
Acquired immune system
cd47
QR1-502
Up-Regulation
Infectious Diseases
Receptors
Pattern Recognition

030220 oncology & carcinogenesis
Cytokines
Female
medicine.symptom
Infection
Research Article
Knockout
CD47 Antigen
Inflammation
Pattern Recognition
Biology
pathogen recognition receptors
Microbiology
Host-Microbe Biology
Cell Line
Immunomodulation
Vaccine Related
Betacoronavirus
03 medical and health sciences
Immune system
Antigen
Cell Line
Tumor

Biodefense
Virology
medicine
Animals
Humans
immune checkpoint
030304 developmental biology
Innate immune system
SARS-CoV-2
Prevention
Inflammatory and immune system
Immunity
Mycobacterium tuberculosis
Immunotherapy
Immunity
Innate

Immune checkpoint
Mice
Inbred C57BL

Emerging Infectious Diseases
Good Health and Well Being
A549 Cells
Immunology
Immunization
Zdroj: mBio
mBio, Vol 11, Iss 3, p e01293-20 (2020)
mBio, vol 11, iss 3
mBio, Vol 11, Iss 3 (2020)
Popis: Immune responses to infectious agents are initiated when a pathogen or its components bind to pattern recognition receptors (PRRs). PRR binding sets off a cascade of events that activates immune responses. We now show that, in addition to activating immune responses, PRR signaling also initiates an immunosuppressive response, probably to limit inflammation. The importance of the current findings is that blockade of immunomodulatory signaling, which is mediated by the upregulation of the CD47 molecule, can lead to enhanced immune responses to any pathogen that triggers PRR signaling. Since most or all pathogens trigger PRRs, CD47 blockade could be used to speed up and strengthen both innate and adaptive immune responses when medically indicated. Such immunotherapy could be done without a requirement for knowing the HLA type of the individual, the specific antigens of the pathogen, or, in the case of bacterial infections, the antimicrobial resistance profile.
It is well understood that the adaptive immune response to infectious agents includes a modulating suppressive component as well as an activating component. We now show that the very early innate response also has an immunosuppressive component. Infected cells upregulate the CD47 “don’t eat me” signal, which slows the phagocytic uptake of dying and viable cells as well as downstream antigen-presenting cell (APC) functions. A CD47 mimic that acts as an essential virulence factor is encoded by all poxviruses, but CD47 expression on infected cells was found to be upregulated even by pathogens, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), that encode no mimic. CD47 upregulation was revealed to be a host response induced by the stimulation of both endosomal and cytosolic pathogen recognition receptors (PRRs). Furthermore, proinflammatory cytokines, including those found in the plasma of hepatitis C patients, upregulated CD47 on uninfected dendritic cells, thereby linking innate modulation with downstream adaptive immune responses. Indeed, results from antibody-mediated CD47 blockade experiments as well as CD47 knockout mice revealed an immunosuppressive role for CD47 during infections with lymphocytic choriomeningitis virus and Mycobacterium tuberculosis. Since CD47 blockade operates at the level of pattern recognition receptors rather than at a pathogen or antigen-specific level, these findings identify CD47 as a novel potential immunotherapeutic target for the enhancement of immune responses to a broad range of infectious agents.
Databáze: OpenAIRE