Cargando…

Influenza Transmission in the Mother-Infant Dyad Leads to Severe Disease, Mammary Gland Infection, and Pathogenesis by Regulating Host Responses

Seasonal influenza viruses are typically restricted to the human upper respiratory tract whereas influenza viruses with greater pathogenic potential often also target extra-pulmonary organs. Infants, pregnant women, and breastfeeding mothers are highly susceptible to severe respiratory disease follo...

Descripción completa

Detalles Bibliográficos
Autores principales: Paquette, Stéphane G., Banner, David, Huang, Stephen S. H., Almansa, Raquel, Leon, Alberto, Xu, Luoling, Bartoszko, Jessica, Kelvin, David J., Kelvin, Alyson A.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4598190/
https://www.ncbi.nlm.nih.gov/pubmed/26448646
http://dx.doi.org/10.1371/journal.ppat.1005173
_version_ 1782394049951432704
author Paquette, Stéphane G.
Banner, David
Huang, Stephen S. H.
Almansa, Raquel
Leon, Alberto
Xu, Luoling
Bartoszko, Jessica
Kelvin, David J.
Kelvin, Alyson A.
author_facet Paquette, Stéphane G.
Banner, David
Huang, Stephen S. H.
Almansa, Raquel
Leon, Alberto
Xu, Luoling
Bartoszko, Jessica
Kelvin, David J.
Kelvin, Alyson A.
author_sort Paquette, Stéphane G.
collection PubMed
description Seasonal influenza viruses are typically restricted to the human upper respiratory tract whereas influenza viruses with greater pathogenic potential often also target extra-pulmonary organs. Infants, pregnant women, and breastfeeding mothers are highly susceptible to severe respiratory disease following influenza virus infection but the mechanisms of disease severity in the mother-infant dyad are poorly understood. Here we investigated 2009 H1N1 influenza virus infection and transmission in breastfeeding mothers and infants utilizing our developed infant-mother ferret influenza model. Infants acquired severe disease and mortality following infection. Transmission of the virus from infants to mother ferrets led to infection in the lungs and mother mortality. Live virus was also found in mammary gland tissue and expressed milk of the mothers which eventually led to milk cessation. Histopathology showed destruction of acini glandular architecture with the absence of milk. The virus was localized in mammary epithelial cells of positive glands. To understand the molecular mechanisms of mammary gland infection, we performed global transcript analysis which showed downregulation of milk production genes such as Prolactin and increased breast involution pathways indicated by a STAT5 to STAT3 signaling shift. Genes associated with cancer development were also significantly increased including JUN, FOS and M2 macrophage markers. Immune responses within the mammary gland were characterized by decreased lymphocyte-associated genes CD3e, IL2Ra, CD4 with IL1β upregulation. Direct inoculation of H1N1 into the mammary gland led to infant respiratory infection and infant mortality suggesting the influenza virus was able to replicate in mammary tissue and transmission is possible through breastfeeding. In vitro infection studies with human breast cells showed susceptibility to H1N1 virus infection. Together, we have shown that the host-pathogen interactions of influenza virus infection in the mother-infant dyad initiate immunological and oncogenic signaling cascades within the mammary gland. These findings suggest the mammary gland may have a greater role in infection and immunity than previously thought.
format Online
Article
Text
id pubmed-4598190
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-45981902015-10-20 Influenza Transmission in the Mother-Infant Dyad Leads to Severe Disease, Mammary Gland Infection, and Pathogenesis by Regulating Host Responses Paquette, Stéphane G. Banner, David Huang, Stephen S. H. Almansa, Raquel Leon, Alberto Xu, Luoling Bartoszko, Jessica Kelvin, David J. Kelvin, Alyson A. PLoS Pathog Research Article Seasonal influenza viruses are typically restricted to the human upper respiratory tract whereas influenza viruses with greater pathogenic potential often also target extra-pulmonary organs. Infants, pregnant women, and breastfeeding mothers are highly susceptible to severe respiratory disease following influenza virus infection but the mechanisms of disease severity in the mother-infant dyad are poorly understood. Here we investigated 2009 H1N1 influenza virus infection and transmission in breastfeeding mothers and infants utilizing our developed infant-mother ferret influenza model. Infants acquired severe disease and mortality following infection. Transmission of the virus from infants to mother ferrets led to infection in the lungs and mother mortality. Live virus was also found in mammary gland tissue and expressed milk of the mothers which eventually led to milk cessation. Histopathology showed destruction of acini glandular architecture with the absence of milk. The virus was localized in mammary epithelial cells of positive glands. To understand the molecular mechanisms of mammary gland infection, we performed global transcript analysis which showed downregulation of milk production genes such as Prolactin and increased breast involution pathways indicated by a STAT5 to STAT3 signaling shift. Genes associated with cancer development were also significantly increased including JUN, FOS and M2 macrophage markers. Immune responses within the mammary gland were characterized by decreased lymphocyte-associated genes CD3e, IL2Ra, CD4 with IL1β upregulation. Direct inoculation of H1N1 into the mammary gland led to infant respiratory infection and infant mortality suggesting the influenza virus was able to replicate in mammary tissue and transmission is possible through breastfeeding. In vitro infection studies with human breast cells showed susceptibility to H1N1 virus infection. Together, we have shown that the host-pathogen interactions of influenza virus infection in the mother-infant dyad initiate immunological and oncogenic signaling cascades within the mammary gland. These findings suggest the mammary gland may have a greater role in infection and immunity than previously thought. Public Library of Science 2015-10-08 /pmc/articles/PMC4598190/ /pubmed/26448646 http://dx.doi.org/10.1371/journal.ppat.1005173 Text en © 2015 Paquette et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Paquette, Stéphane G.
Banner, David
Huang, Stephen S. H.
Almansa, Raquel
Leon, Alberto
Xu, Luoling
Bartoszko, Jessica
Kelvin, David J.
Kelvin, Alyson A.
Influenza Transmission in the Mother-Infant Dyad Leads to Severe Disease, Mammary Gland Infection, and Pathogenesis by Regulating Host Responses
title Influenza Transmission in the Mother-Infant Dyad Leads to Severe Disease, Mammary Gland Infection, and Pathogenesis by Regulating Host Responses
title_full Influenza Transmission in the Mother-Infant Dyad Leads to Severe Disease, Mammary Gland Infection, and Pathogenesis by Regulating Host Responses
title_fullStr Influenza Transmission in the Mother-Infant Dyad Leads to Severe Disease, Mammary Gland Infection, and Pathogenesis by Regulating Host Responses
title_full_unstemmed Influenza Transmission in the Mother-Infant Dyad Leads to Severe Disease, Mammary Gland Infection, and Pathogenesis by Regulating Host Responses
title_short Influenza Transmission in the Mother-Infant Dyad Leads to Severe Disease, Mammary Gland Infection, and Pathogenesis by Regulating Host Responses
title_sort influenza transmission in the mother-infant dyad leads to severe disease, mammary gland infection, and pathogenesis by regulating host responses
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4598190/
https://www.ncbi.nlm.nih.gov/pubmed/26448646
http://dx.doi.org/10.1371/journal.ppat.1005173
work_keys_str_mv AT paquettestephaneg influenzatransmissioninthemotherinfantdyadleadstoseverediseasemammaryglandinfectionandpathogenesisbyregulatinghostresponses
AT bannerdavid influenzatransmissioninthemotherinfantdyadleadstoseverediseasemammaryglandinfectionandpathogenesisbyregulatinghostresponses
AT huangstephensh influenzatransmissioninthemotherinfantdyadleadstoseverediseasemammaryglandinfectionandpathogenesisbyregulatinghostresponses
AT almansaraquel influenzatransmissioninthemotherinfantdyadleadstoseverediseasemammaryglandinfectionandpathogenesisbyregulatinghostresponses
AT leonalberto influenzatransmissioninthemotherinfantdyadleadstoseverediseasemammaryglandinfectionandpathogenesisbyregulatinghostresponses
AT xuluoling influenzatransmissioninthemotherinfantdyadleadstoseverediseasemammaryglandinfectionandpathogenesisbyregulatinghostresponses
AT bartoszkojessica influenzatransmissioninthemotherinfantdyadleadstoseverediseasemammaryglandinfectionandpathogenesisbyregulatinghostresponses
AT kelvindavidj influenzatransmissioninthemotherinfantdyadleadstoseverediseasemammaryglandinfectionandpathogenesisbyregulatinghostresponses
AT kelvinalysona influenzatransmissioninthemotherinfantdyadleadstoseverediseasemammaryglandinfectionandpathogenesisbyregulatinghostresponses