학술논문

The phosphatase CD148 promotes airway hyperresponsiveness through SRC family kinases.
Document Type
article
Source
The Journal of clinical investigation. 123(5)
Subject
Lung
Bronchi
Trachea
Myocytes
Smooth Muscle
Animals
Mice
Inbred BALB C
Mice
Inbred C57BL
Mice
Transgenic
Mice
Asthma
Inflammation
src-Family Kinases
Ovalbumin
Signal Transduction
Gene Deletion
Cell Lineage
Female
Male
Receptor-Like Protein Tyrosine Phosphatases
Class 3
Myocytes
Smooth Muscle
Inbred BALB C
Inbred C57BL
Transgenic
Receptor-Like Protein Tyrosine Phosphatases
Class 3
Medical and Health Sciences
Immunology
Language
Abstract
Increased airway smooth muscle (ASM) contractility and the development of airway hyperresponsiveness (AHR) are cardinal features of asthma, but the signaling pathways that promote these changes are poorly understood. Tyrosine phosphorylation is tightly regulated by the opposing actions of protein tyrosine kinases and phosphatases, but little is known about whether tyrosine phosphatases influence AHR. Here, we demonstrate that genetic inactivation of receptor-like protein tyrosine phosphatase J (Ptprj), which encodes CD148, protected mice from the development of increased AHR in two different asthma models. Surprisingly, CD148 deficiency minimally affected the inflammatory response to allergen, but significantly altered baseline pulmonary resistance. Mice specifically lacking CD148 in smooth muscle had decreased AHR, and the frequency of calcium oscillations in CD148-deficient ASM was substantially attenuated, suggesting that signaling pathway alterations may underlie ASM contractility. Biochemical analysis of CD148-deficient ASM revealed hyperphosphorylation of the C-terminal inhibitory tyrosine of SRC family kinases (SFKs), implicating CD148 as a critical positive regulator of SFK signaling in ASM. The effect of CD148 deficiency on ASM contractility could be mimicked by treatment of both mouse trachea and human bronchi with specific SFK inhibitors. Our studies identify CD148 and the SFKs it regulates in ASM as potential targets for the treatment of AHR.