[HTML][HTML] CD4+ cells regulate fibrosis and lymphangiogenesis in response to lymphatic fluid stasis

JC Zampell, A Yan, S Elhadad, T Avraham, E Weitman… - PloS one, 2012 - journals.plos.org
JC Zampell, A Yan, S Elhadad, T Avraham, E Weitman, BJ Mehrara
PloS one, 2012journals.plos.org
Introduction Lymphedema is a chronic disorder that occurs commonly after lymph node
removal for cancer treatment and is characterized by swelling, fibrosis, inflammation, and
adipose deposition. Although previous histological studies have investigated inflammatory
changes that occur in lymphedema, the precise cellular make up of the inflammatory
infiltrate remains unknown. It is also unclear if this inflammatory response plays a causal role
in the pathology of lymphedema. The purpose of this study was therefore to characterize the …
Introduction
Lymphedema is a chronic disorder that occurs commonly after lymph node removal for cancer treatment and is characterized by swelling, fibrosis, inflammation, and adipose deposition. Although previous histological studies have investigated inflammatory changes that occur in lymphedema, the precise cellular make up of the inflammatory infiltrate remains unknown. It is also unclear if this inflammatory response plays a causal role in the pathology of lymphedema. The purpose of this study was therefore to characterize the inflammatory response to lymphatic stasis and determine if these responses are necessary for the pathological changes that occur in lymphedema.
Methods
We used mouse-tail lymphedema and axillary lymph node dissection (ANLD) models in order to study tissue inflammatory changes. Single cell suspensions were created and analyzed using multi-color flow cytometry to identify individual cell types. We utilized antibody depletion techniques to analyze the causal role of CD4+, CD8+, and CD25+ cells in the regulation of inflammation, fibrosis, adipose deposition, and lymphangiogenesis.
Results
Lymphedema in the mouse-tail resulted in a mixed inflammatory cell response with significant increases in T-helper, T-regulatory, neutrophils, macrophages, and dendritic cell populations. Interestingly, we found that ALND resulted in significant increases in T-helper cells suggesting that these adaptive immune responses precede changes in macrophage and dendritic cell infiltration. In support of this we found that depletion of CD4+, but not CD8 or CD25+ cells, significantly decreased tail lymphedema, inflammation, fibrosis, and adipose deposition. In addition, depletion of CD4+ cells significantly increased lymphangiogenesis both in our tail model and also in an inflammatory lymphangiogenesis model.
Conclusions
Lymphedema and lymphatic stasis result in CD4+ cell inflammation and infiltration of mature T-helper cells. Loss of CD4+ but not CD8+ or CD25+ cell inflammation markedly decreases the pathological changes associated with lymphedema. In addition, CD4+ cells regulate lymphangiogenesis during wound repair and inflammatory lymphangiogenesis.
PLOS