Your browser doesn't support javascript.
loading
Endometrial stromal cells exhibit a distinct phenotypic and immunomodulatory profile.
Queckbörner, Suzanna; Syk Lundberg, Elisabeth; Gemzell-Danielsson, Kristina; Davies, Lindsay C.
Afiliación
  • Queckbörner S; Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, and Karolinska University Hospital, S-171 64, Solna, Sweden. suzanna.queckborner@ki.se.
  • Syk Lundberg E; Department of Clinical Genetics, Karolinska University Hospital, S-171 76, Stockholm, Sweden.
  • Gemzell-Danielsson K; Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet, and Karolinska University Hospital, S-171 64, Solna, Sweden.
  • Davies LC; Department of Laboratory Medicine, Karolinska Institutet, S-141 52, Huddinge, Sweden.
Stem Cell Res Ther ; 11(1): 15, 2020 01 06.
Article en En | MEDLINE | ID: mdl-31907034
ABSTRACT

BACKGROUND:

In Asherman's syndrome (AS), intrauterine scarring and fibrotic adhesions lead to menstrual disorders, pregnancy loss, or infertility. A few clinical trials have piloted cell therapy to overcome AS. Understanding the role of the stromal compartment in endometrial regeneration remains poorly understood. We hypothesize that endometrial stromal cells (eSCs) represent a relevant cell population to establish novel cell-based therapeutics for endometrial disorders. The aim of this study was to characterize eSCs and evaluate their immune-cell interactions.

METHODS:

eSCs were isolated from healthy donors, during the proliferative stage of the menstrual cycle. Cells were characterized for expression of mesenchymal stromal cell (MSC) markers and assessed for their tumorigenic potential. eSCs were co-cultured with interferon γ and tumor necrosis factor α, and cell surface expression of their respective receptors and human leukocyte antigen (HLA) I and II determined by flow cytometry. Secreted levels of key immunomodulatory factors were established. eSCs were cultured with activated peripheral blood mononuclear cells, and T cell differentiation and proliferation determined.

RESULTS:

eSCs demonstrated an MSC surface phenotype and exhibited multipotency. Expanded eSCs retained chromosomal stability and demonstrated no tumorigenicity. Upon stimulation, eSCs licensed to an anti-inflammatory phenotype with upregulated secretion of immunomodulatory factors. Stimulated eSCs did not express HLA class II. eSCs suppressed the proliferation and activation of CD4+ T cells, with the eSC secretome further downregulating central memory T cells and upregulating effector memory (EM) cells.

CONCLUSIONS:

Differential responsiveness to inflammation by eSCs, compared to other MSC sources, demonstrates the need to understand the specific functional effects of individual stromal cell sources. A lack of HLA class II and triggering of EM T cell differentiation strongly links to innate in vivo roles of eSCs in tissue repair and immune tolerance during pregnancy. We conclude that eSCs may be an ideal cell therapy candidate for endometrial disorders.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Células del Estroma / Inmunomodulación Límite: Adult / Female / Humans Idioma: En Revista: Stem Cell Res Ther Año: 2020 Tipo del documento: Article País de afiliación: Suecia

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Células del Estroma / Inmunomodulación Límite: Adult / Female / Humans Idioma: En Revista: Stem Cell Res Ther Año: 2020 Tipo del documento: Article País de afiliación: Suecia