Your browser doesn't support javascript.
loading
The Role of Macrophage Migration Inhibitory Factor in Adipose-Derived Stem Cells Under Hypoxia.
Hofmann, Elena; Soppert, Josefin; Ruhl, Tim; Gousopoulos, Epameinondas; Gerra, Simona; Storti, Gabriele; Tian, Yuan; Brandhofer, Markus; Schweizer, Riccardo; Song, Seung-Yong; Lindenblatt, Nicole; Pallua, Norbert; Bernhagen, Jürgen; Kim, Bong-Sung.
Afiliación
  • Hofmann E; Department of Plastic Surgery and Hand Surgery-Burn Center, University Hospital RWTH Aachen, Aachen, Germany.
  • Soppert J; Institute of Biochemistry and Molecular Cell Biology, University Hospital RWTH Aachen, Aachen, Germany.
  • Ruhl T; Institute of Biochemistry and Molecular Cell Biology, University Hospital RWTH Aachen, Aachen, Germany.
  • Gousopoulos E; Institute for Molecular Cardiovascular Research, University Hospital RWTH Aachen, Aachen, Germany.
  • Gerra S; Department of Intensive Care and Intermediate Care, University Hospital RWTH Aachen, Aachen, Germany.
  • Storti G; Department of Plastic Surgery and Hand Surgery-Burn Center, University Hospital RWTH Aachen, Aachen, Germany.
  • Tian Y; Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Zurich, Switzerland.
  • Brandhofer M; Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig Maximilian University of Munich (LMU), Munich, Germany.
  • Schweizer R; Plastic and Reconstructive Surgery, Department of Surgical Sciences, University of Rome "Tor Vergata", Rome, Italy.
  • Song SY; Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig Maximilian University of Munich (LMU), Munich, Germany.
  • Lindenblatt N; Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig Maximilian University of Munich (LMU), Munich, Germany.
  • Pallua N; Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Zurich, Switzerland.
  • Bernhagen J; Department of Plastic and Reconstructive Surgery, Yonsei University College of Medicine, Seoul, South Korea.
  • Kim BS; Department of Plastic Surgery and Hand Surgery, University Hospital of Zürich, Zurich, Switzerland.
Front Physiol ; 12: 638448, 2021.
Article en En | MEDLINE | ID: mdl-34366876
ABSTRACT

Background:

Adipose-derived stem cells (ASCs) are multipotent mesenchymal stem cells characterized by their strong regenerative potential and low oxygen consumption. Macrophage migration inhibitory factor (MIF) is a multifunctional chemokine-like cytokine that is involved in tissue hypoxia. MIF is not only a major immunomodulator but also is highly expressed in adipose tissue such as subcutaneous adipose tissue of chronic non-healing wounds. In the present study, we investigated the effect of hypoxia on MIF in ASCs isolated from healthy versus inflamed adipose tissue.

Methods:

Human ASCs were harvested from 17 patients (11 healthy adipose tissue samples, six specimens from chronic non-healing wounds). ASCs were treated in a hypoxia chamber at <1% oxygen. ASC viability, MIF secretion as well as expression levels of MIF, its receptor CD74, hypoxia-inducible transcription factor-1α (HIF-1α) and activation of the AKT and ERK signaling pathways were analyzed. The effect of recombinant MIF on the viability of ASCs was determined. Finally, the effect of MIF on the viability and production capacity of ASCs to produce the inflammatory cytokines tumor necrosis factor (TNF), interleukin (IL)-6, and IL-1ß was determined upon treatment with recombinant MIF and/or a blocking MIF antibody.

Results:

Hypoxic treatment inhibited proliferation of ASCs derived from healthy or chronic non-healing wounds. ASCs from healthy adipose tissue samples were characterized by a low degree of MIF secretion during hypoxic challenge. In contrast, in ASCs from adipose tissue samples of chronic non-healing wounds, secretion and expression of MIF and CD74 expression were significantly elevated under hypoxia. This was accompanied by enhanced ERK signaling, while AKT signaling was not altered. Recombinant MIF did stimulate HIF-1α expression under hypoxia as well as AKT and ERK phosphorylation, while no effect on ASC viability was observed. Recombinant MIF significantly reduced the secretion of IL-1ß under hypoxia and normoxia, and neutralizing MIF-antibodies diminished TNF-α and IL-1ß release in hypoxic ASCs.

Conclusions:

Collectively, MIF did not affect the viability of ASCs from neither healthy donor site nor chronic wounds. Our results, however, suggest that MIF has an impact on the wound environment by modulating inflammatory factors such as IL-1ß.
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Idioma: En Revista: Front Physiol Año: 2021 Tipo del documento: Article País de afiliación: Alemania

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Idioma: En Revista: Front Physiol Año: 2021 Tipo del documento: Article País de afiliación: Alemania