[HTML][HTML] Real-time monitoring of tumorigenesis, dissemination, & drug response in a preclinical model of lymphangioleiomyomatosis/tuberous sclerosis complex

F Liu, EP Lunsford, J Tong, Y Ashitate, SL Gibbs, J Yu… - PloS one, 2012 - journals.plos.org
F Liu, EP Lunsford, J Tong, Y Ashitate, SL Gibbs, J Yu, HS Choi, EP Henske, JV Frangioni
PloS one, 2012journals.plos.org
Background TSC2-deficient cells can proliferate in the lungs, kidneys, and other organs
causing devastating progressive multisystem disorders such as lymphangioleiomyomatosis
(LAM) and tuberous sclerosis complex (TSC). Preclinical models utilizing LAM patient-
derived cells have been difficult to establish. We developed a novel animal model system to
study the molecular mechanisms of TSC/LAM pathogenesis and tumorigenesis and provide
a platform for drug testing. Methods and Findings TSC2-deficient human cells, derived from …
Background
TSC2-deficient cells can proliferate in the lungs, kidneys, and other organs causing devastating progressive multisystem disorders such as lymphangioleiomyomatosis (LAM) and tuberous sclerosis complex (TSC). Preclinical models utilizing LAM patient-derived cells have been difficult to establish. We developed a novel animal model system to study the molecular mechanisms of TSC/LAM pathogenesis and tumorigenesis and provide a platform for drug testing.
Methods and Findings
TSC2-deficient human cells, derived from the angiomyolipoma of a LAM patient, were engineered to co-express both sodium-iodide symporter (NIS) and green fluorescent protein (GFP). Cells were inoculated intraparenchymally, intravenously, or intratracheally into athymic NCr nu/nu mice and cells were tracked and quantified using single photon emission computed tomography (SPECT) and computed tomography (CT). Surprisingly, TSC2-deficient cells administered intratracheally resulted in rapid dissemination to lymph node basins throughout the body, and histopathological changes in the lung consistent with LAM. Estrogen was found to be permissive for tumor growth and dissemination. Rapamycin inhibited tumor growth, but tumors regrew after the drug treatment was withdrawn.
Conclusions
We generated homogeneous NIS/GFP co-expressing TSC2-deficient, patient-derived cells that can proliferate and migrate in vivo after intratracheal instillation. Although the animal model we describe has some limitations, we demonstrate that systemic tumors formed from TSC2-deficient cells can be monitored and quantified noninvasively over time using SPECT/CT, thus providing a much needed model system for in vivo drug testing and mechanistic studies of TSC2-deficient cells and their related clinical syndromes.
PLOS