Background: In humans, interferon-a treatment for chronic viral hepatitis is a well-recognized clinical model for inflammationinduced depression, but the molecular mechanisms underlying these effects are not clear. Following peripheral administration in rodents, interferon-a induces signal transducer and activator of transcription-1 (STAT1) within the hippocampus and disrupts hippocampal neurogenesis. Methods: We used the human hippocampal progenitor cell line HPC0A07/03C to evaluate the effects of 2 concentrations of interferon-a, similar to those observed in human serum during its therapeutic use (500 pg/mL and 5000 pg/mL), on neurogenesis and apoptosis. Results: Both concentrations of interferon-a decreased hippocampal neurogenesis, with the high concentration also increasing apoptosis. Moreover, interferon-a increased the expression of interferon-stimulated gene 15 (ISG15), ubiquitin-specific peptidase 18 (USP18), and interleukin-6 (IL-6) via activation of STAT1. Like interferon-a, co-treatment with a combination of ISG15, USP18, and IL-6 was able to reduce neurogenesis and enhance apoptosis via further downstream activation of STAT1. Further experiments showed that ISG15 and USP18 mediated the interferon-a-induced reduction in neurogenesis (potentially through upregulation of the ISGylation-related proteins UBA7, UBE2L6, and HERC5), while IL-6 mediated the interferon- a-induced increase in apoptosis (potentially through downregulation of aquaporin 4). Using transcriptomic analyses, we showed that interferon-a regulated pathways involved in oxidative stress and immune response (e.g., Nuclear Factor (erythroid-derived 2)-like 2 [Nrf2] and interferon regulatory factor [IRF] signaling pathway), neuronal formation (e.g., CAMP response element-binding protein [CREB] signaling), and cell death regulation (e.g., tumor protein(p)53 signaling). Conclusions: We identify novel molecular mechanisms mediating the effects of interferon-a on the human hippocampus potentially involved in inflammation-induced neuropsychiatric symptoms.

Interferon-alpha reduces human hippocampal neurogenesis and increases apoptosis via activation of distinct STAT1-dependent mechanisms / A. Borsini, A. Cattaneo, C. Malpighi, S. Thuret, N.A. Harrison, P.A. Zunszain, C.M. Pariante. - In: INTERNATIONAL JOURNAL OF NEUROPSYCHOPHARMACOLOGY. - ISSN 1461-1457. - 21:2(2018 Feb), pp. 187-200. [10.1093/ijnp/pyx083]

Interferon-alpha reduces human hippocampal neurogenesis and increases apoptosis via activation of distinct STAT1-dependent mechanisms

A. Cattaneo;C. Malpighi;
2018

Abstract

Background: In humans, interferon-a treatment for chronic viral hepatitis is a well-recognized clinical model for inflammationinduced depression, but the molecular mechanisms underlying these effects are not clear. Following peripheral administration in rodents, interferon-a induces signal transducer and activator of transcription-1 (STAT1) within the hippocampus and disrupts hippocampal neurogenesis. Methods: We used the human hippocampal progenitor cell line HPC0A07/03C to evaluate the effects of 2 concentrations of interferon-a, similar to those observed in human serum during its therapeutic use (500 pg/mL and 5000 pg/mL), on neurogenesis and apoptosis. Results: Both concentrations of interferon-a decreased hippocampal neurogenesis, with the high concentration also increasing apoptosis. Moreover, interferon-a increased the expression of interferon-stimulated gene 15 (ISG15), ubiquitin-specific peptidase 18 (USP18), and interleukin-6 (IL-6) via activation of STAT1. Like interferon-a, co-treatment with a combination of ISG15, USP18, and IL-6 was able to reduce neurogenesis and enhance apoptosis via further downstream activation of STAT1. Further experiments showed that ISG15 and USP18 mediated the interferon-a-induced reduction in neurogenesis (potentially through upregulation of the ISGylation-related proteins UBA7, UBE2L6, and HERC5), while IL-6 mediated the interferon- a-induced increase in apoptosis (potentially through downregulation of aquaporin 4). Using transcriptomic analyses, we showed that interferon-a regulated pathways involved in oxidative stress and immune response (e.g., Nuclear Factor (erythroid-derived 2)-like 2 [Nrf2] and interferon regulatory factor [IRF] signaling pathway), neuronal formation (e.g., CAMP response element-binding protein [CREB] signaling), and cell death regulation (e.g., tumor protein(p)53 signaling). Conclusions: We identify novel molecular mechanisms mediating the effects of interferon-a on the human hippocampus potentially involved in inflammation-induced neuropsychiatric symptoms.
English
Apoptosis; Depression; Inflammation; Interferon-alpha; Neurogenesis; Apoptosis; Cell Line; Hippocampus; Humans; Inflammation; Interferon-alpha; Neurogenesis; STAT1 Transcription Factor; Stem Cells
Settore BIO/14 - Farmacologia
Articolo
Esperti anonimi
Pubblicazione scientifica
feb-2018
Oxford University Press
21
2
187
200
14
Pubblicato
Periodico con rilevanza internazionale
scopus
Aderisco
info:eu-repo/semantics/article
Interferon-alpha reduces human hippocampal neurogenesis and increases apoptosis via activation of distinct STAT1-dependent mechanisms / A. Borsini, A. Cattaneo, C. Malpighi, S. Thuret, N.A. Harrison, P.A. Zunszain, C.M. Pariante. - In: INTERNATIONAL JOURNAL OF NEUROPSYCHOPHARMACOLOGY. - ISSN 1461-1457. - 21:2(2018 Feb), pp. 187-200. [10.1093/ijnp/pyx083]
open
Prodotti della ricerca::01 - Articolo su periodico
7
262
Article (author)
Periodico con Impact Factor
A. Borsini, A. Cattaneo, C. Malpighi, S. Thuret, N.A. Harrison, P.A. Zunszain, C.M. Pariante
File in questo prodotto:
File Dimensione Formato  
Borsini 2017.pdf

accesso aperto

Tipologia: Publisher's version/PDF
Dimensione 1.28 MB
Formato Adobe PDF
1.28 MB Adobe PDF Visualizza/Apri
Pubblicazioni consigliate

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/2434/732387
Citazioni
  • ???jsp.display-item.citation.pmc??? 46
  • Scopus 74
  • ???jsp.display-item.citation.isi??? 70
social impact