[HTML][HTML] Genetically reprogrammed, liver-derived insulin-producing cells are glucose-responsive, but susceptible to autoimmune destruction in settings of murine …

DQ Tang, L Shun, V Koya, Y Sun, Q Wang… - American Journal of …, 2013 - ncbi.nlm.nih.gov
DQ Tang, L Shun, V Koya, Y Sun, Q Wang, H Wang, SW Li, Y Sun, DL Purich, C Zhang…
American Journal of Translational Research, 2013ncbi.nlm.nih.gov
Many previous studies demonstrate that hepatocytes can be reprogrammed into insulin-
producing cells (IPCs) utilizing viral vector-mediated delivery of pancreatic transcription
factors (PTFs). However, whether these liver-derived IPCs are susceptible to autoimmune
attack in animal models of type 1 diabetes remains unclear, in part due to the
immunogenicity of the viral vectors used to introduce PTF genes. Adeno-associated virus
serotype 2 vector-expressing Pdx1-VP16 (Pdx1) and Ngn3 were prepared and injected into …
Abstract
Many previous studies demonstrate that hepatocytes can be reprogrammed into insulin-producing cells (IPCs) utilizing viral vector-mediated delivery of pancreatic transcription factors (PTFs). However, whether these liver-derived IPCs are susceptible to autoimmune attack in animal models of type 1 diabetes remains unclear, in part due to the immunogenicity of the viral vectors used to introduce PTF genes. Adeno-associated virus serotype 2 vector-expressing Pdx1-VP16 (Pdx1) and Ngn3 were prepared and injected into the portal vein of streptozotocin (Stz)/diabetic NOD/SCID mice. The presence of glucose-responsive liver-IPCs and their susceptibility to anti-beta cell autoimmunity were assessed by blood glucose levels, insulin content, IPC cell distribution, and intraperitoneal glucose tolerance test following subtotal pancreatectomy (Px) and passive transfer of diabetogenic splenocytes isolated from diabetic female NOD mice. A combination of two PTF genes (Pdx1/Ngn3) effectively reprogrammed liver cells into glucose-responsive IPCs. These IPCs corrected hyperglycemia in Stz/diabetic NOD/SCID mice and maintained normoglycemia following subtotal Px, indicating that liver-derived IPCs could maintain glucose homeostasis. Importantly, we also demonstrated that the glucose-responsive liver–derived IPCs were susceptible to autoimmune destruction by diabetogenic splenocytes, as indicated by progressive elevation in blood glucose levels as well as mixed T-, and B-lymphocytic infiltrates surrounding liver-IPCs 2~ 3 weeks following transferring of diabetogenic splenocytes into NOD/SCID mice, and confirmed by immunohistochemical studies. In conclusion, genetically reprogrammed liver-IPCs, like pancreatic islet beta-cells, are susceptible to autoimmune attack, suggesting that for cell-replacement therapy of treating type 1 diabetes, beta-cell surrogates may require concomitant immunotherapy to avoid autoimmune destruction.
ncbi.nlm.nih.gov