Skip to main content
Fig. 6 | Molecular Brain

Fig. 6

From: The voltage-gated proton channel Hv1 promotes microglia-astrocyte communication and neuropathic pain after peripheral nerve injury

Fig. 6

Hv1 is required for astrocyte-derived IFN-γ to promote neuropathic pain after SNT. A Real-time PCR analysis data showing mRNA expression fold change of glial-derived cytokines in the spinal cord at POD3 after SNT. Data are presented as mean ± SEM, n = 6 runs from 3 sample pools, each sample pool contained 2 mouse samples for each group. *p < 0.05 unpaired t-test. B Representative images of IFN-γ (top panels) and IFN-γ / GFAP merged (bottom panels) immunoreactivity in the ipsilateral dorsal horn in control (a naïve WT sample was shown, left), POD3 SNT-induced WT (left center), and Hv1 KO (center), as well as POD7 SNT-induced WT (right center) and Hv1 KO (right) tissues. Inserts are higher magnification of boxed regions. Scale bar: 50 µm. C Quantitation of IFN-γ immunoreactivity in control as well as POD3 and 7 SNT-treated WT and Hv1 KO mice. Data are presented as mean ± SEM, cells were randomly picked from 3 mice for each group. ***p < 0.001. WT vs. Hv1 KO, unpaired t-test. D Mechanical allodynia in WT and Hv1 KO mice treated with or without neutralizing IFN-γ antibody (anti-IFN-γ). Anti-IFN-γ (1 μg in 5 μl ACSF) was applied intrathecally at POD3 and POD4 after SNT. Anti-IFN-γ intrathecal injection attenuated mechanical allodynia in WT mice when compared to the control group (WT + saline) but not in HV1 KO mice with anti-IFN-γ or saline injection. Data are presented as mean ± SEM. n = 7 mice for each group, **p < 0.01, unpaired t-test

Back to article page