Cargando…

Dendrimer-Based N-Acetyl Cysteine Maternal Therapy Ameliorates Placental Inflammation via Maintenance of M1/M2 Macrophage Recruitment

Intrauterine inflammation (IUI) is the primary cause of spontaneous preterm birth and predisposes neonates to long-term sequelae, including adverse neurological outcomes. N-acetyl-L-cysteine (NAC) is the amino acid L-cysteine derivative and a precursor to the antioxidant glutathione (GSH). NAC is co...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Yang, Na, Quan, Liu, Jin, Liu, Anguo, Oppong, Akosua, Lee, Ji Yeon, Chudnovets, Anna, Lei, Jun, Sharma, Rishi, Kannan, Sujatha, Kannan, Rangaramanujam M., Burd, Irina
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8831692/
https://www.ncbi.nlm.nih.gov/pubmed/35155393
http://dx.doi.org/10.3389/fbioe.2022.819593
_version_ 1784648558077542400
author Liu, Yang
Na, Quan
Liu, Jin
Liu, Anguo
Oppong, Akosua
Lee, Ji Yeon
Chudnovets, Anna
Lei, Jun
Sharma, Rishi
Kannan, Sujatha
Kannan, Rangaramanujam M.
Burd, Irina
author_facet Liu, Yang
Na, Quan
Liu, Jin
Liu, Anguo
Oppong, Akosua
Lee, Ji Yeon
Chudnovets, Anna
Lei, Jun
Sharma, Rishi
Kannan, Sujatha
Kannan, Rangaramanujam M.
Burd, Irina
author_sort Liu, Yang
collection PubMed
description Intrauterine inflammation (IUI) is the primary cause of spontaneous preterm birth and predisposes neonates to long-term sequelae, including adverse neurological outcomes. N-acetyl-L-cysteine (NAC) is the amino acid L-cysteine derivative and a precursor to the antioxidant glutathione (GSH). NAC is commonly used clinically as an antioxidant with anti-inflammatory properties. Poor bioavailability and high protein binding of NAC necessitates the use of high doses resulting in side effects including nausea, vomiting, and gastric disruptions. Therefore, dendrimer-based therapy can specifically target the drug to the cells involved in inflammation, reducing side effects with efficacy at much lower doses than the free drug. Towards development of the new therapies for the treatment of maternal inflammation, we successfully administered dendrimer-based N-Acetyl Cysteine (DNAC) in an animal model of IUI to reduce preterm birth and perinatal inflammatory response. This study explored the associated immune mechanisms of DNAC treatment on placental macrophages following IUI, especially on M1/M2 type macrophage polarization. Our results demonstrated that intraperitoneal maternal DNAC administration significantly reduced the pro-inflammatory cytokine mRNA of Il1β and Nos2, and decreased CD45(+) leukocyte infiltration in the placenta following IUI. Furthermore, we found that DNAC altered placental immune profile by stimulating macrophages to change to the M2 phenotype while decreasing the M1 phenotype, thus suppressing the inflammatory responses in the placenta. Our study provides evidence for DNAC therapy to alleviate IUI via the maintenance of macrophage M1/M2 imbalance in the placenta.
format Online
Article
Text
id pubmed-8831692
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-88316922022-02-12 Dendrimer-Based N-Acetyl Cysteine Maternal Therapy Ameliorates Placental Inflammation via Maintenance of M1/M2 Macrophage Recruitment Liu, Yang Na, Quan Liu, Jin Liu, Anguo Oppong, Akosua Lee, Ji Yeon Chudnovets, Anna Lei, Jun Sharma, Rishi Kannan, Sujatha Kannan, Rangaramanujam M. Burd, Irina Front Bioeng Biotechnol Bioengineering and Biotechnology Intrauterine inflammation (IUI) is the primary cause of spontaneous preterm birth and predisposes neonates to long-term sequelae, including adverse neurological outcomes. N-acetyl-L-cysteine (NAC) is the amino acid L-cysteine derivative and a precursor to the antioxidant glutathione (GSH). NAC is commonly used clinically as an antioxidant with anti-inflammatory properties. Poor bioavailability and high protein binding of NAC necessitates the use of high doses resulting in side effects including nausea, vomiting, and gastric disruptions. Therefore, dendrimer-based therapy can specifically target the drug to the cells involved in inflammation, reducing side effects with efficacy at much lower doses than the free drug. Towards development of the new therapies for the treatment of maternal inflammation, we successfully administered dendrimer-based N-Acetyl Cysteine (DNAC) in an animal model of IUI to reduce preterm birth and perinatal inflammatory response. This study explored the associated immune mechanisms of DNAC treatment on placental macrophages following IUI, especially on M1/M2 type macrophage polarization. Our results demonstrated that intraperitoneal maternal DNAC administration significantly reduced the pro-inflammatory cytokine mRNA of Il1β and Nos2, and decreased CD45(+) leukocyte infiltration in the placenta following IUI. Furthermore, we found that DNAC altered placental immune profile by stimulating macrophages to change to the M2 phenotype while decreasing the M1 phenotype, thus suppressing the inflammatory responses in the placenta. Our study provides evidence for DNAC therapy to alleviate IUI via the maintenance of macrophage M1/M2 imbalance in the placenta. Frontiers Media S.A. 2022-01-28 /pmc/articles/PMC8831692/ /pubmed/35155393 http://dx.doi.org/10.3389/fbioe.2022.819593 Text en Copyright © 2022 Liu, Na, Liu, Liu, Oppong, Lee, Chudnovets, Lei, Sharma, Kannan, Kannan and Burd. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Bioengineering and Biotechnology
Liu, Yang
Na, Quan
Liu, Jin
Liu, Anguo
Oppong, Akosua
Lee, Ji Yeon
Chudnovets, Anna
Lei, Jun
Sharma, Rishi
Kannan, Sujatha
Kannan, Rangaramanujam M.
Burd, Irina
Dendrimer-Based N-Acetyl Cysteine Maternal Therapy Ameliorates Placental Inflammation via Maintenance of M1/M2 Macrophage Recruitment
title Dendrimer-Based N-Acetyl Cysteine Maternal Therapy Ameliorates Placental Inflammation via Maintenance of M1/M2 Macrophage Recruitment
title_full Dendrimer-Based N-Acetyl Cysteine Maternal Therapy Ameliorates Placental Inflammation via Maintenance of M1/M2 Macrophage Recruitment
title_fullStr Dendrimer-Based N-Acetyl Cysteine Maternal Therapy Ameliorates Placental Inflammation via Maintenance of M1/M2 Macrophage Recruitment
title_full_unstemmed Dendrimer-Based N-Acetyl Cysteine Maternal Therapy Ameliorates Placental Inflammation via Maintenance of M1/M2 Macrophage Recruitment
title_short Dendrimer-Based N-Acetyl Cysteine Maternal Therapy Ameliorates Placental Inflammation via Maintenance of M1/M2 Macrophage Recruitment
title_sort dendrimer-based n-acetyl cysteine maternal therapy ameliorates placental inflammation via maintenance of m1/m2 macrophage recruitment
topic Bioengineering and Biotechnology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8831692/
https://www.ncbi.nlm.nih.gov/pubmed/35155393
http://dx.doi.org/10.3389/fbioe.2022.819593
work_keys_str_mv AT liuyang dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment
AT naquan dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment
AT liujin dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment
AT liuanguo dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment
AT oppongakosua dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment
AT leejiyeon dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment
AT chudnovetsanna dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment
AT leijun dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment
AT sharmarishi dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment
AT kannansujatha dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment
AT kannanrangaramanujamm dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment
AT burdirina dendrimerbasednacetylcysteinematernaltherapyamelioratesplacentalinflammationviamaintenanceofm1m2macrophagerecruitment