Transcriptional signature of islet neogenesis-associated protein peptide-treated rat pancreatic islets reveals induction of novel long non-coding RNAs

Front Endocrinol (Lausanne). 2023 Sep 29:14:1226615. doi: 10.3389/fendo.2023.1226615. eCollection 2023.

Abstract

Background: Diabetes mellitus is characterized by chronic hyperglycemia with loss of β-cell function and mass. An attractive therapeutic approach to treat patients with diabetes in a non-invasive way is to harness the innate regenerative potential of the pancreas. The Islet Neogenesis-Associated Protein pentadecapeptide (INGAP-PP) has been shown to induce β-cell regeneration and improve their function in rodents. To investigate its possible mechanism of action, we report here the global transcriptional effects induced by the short-term INGAP-PP in vitro treatment of adult rat pancreatic islets.

Methods and findings: Rat pancreatic islets were cultured in vitro in the presence of INGAP-PP for 4 days, and RNA-seq was generated from triplicate treated and control islet samples. We performed a de novo rat gene annotation based on the alignment of RNA-seq reads. The list of INGAP-PP-regulated genes was integrated with epigenomic data. Using the new gene annotation generated in this work, we quantified RNA-seq data profiled in INS-1 cells treated with IL1β, IL1β+Calcipotriol (a vitamin D agonist) or vehicle, and single-cell RNA-seq data profiled in rat pancreatic islets. We found 1,669 differentially expressed genes by INGAP-PP treatment, including dozens of previously unannotated rat transcripts. Genes differentially expressed by the INGAP-PP treatment included a subset of upregulated transcripts that are associated with vitamin D receptor activation. Supported by epigenomic and single-cell RNA-seq data, we identified 9 previously unannotated long noncoding RNAs (lncRNAs) upregulated by INGAP-PP, some of which are also differentially regulated by IL1β and vitamin D in β-cells. These include Ri-lnc1, which is enriched in mature β-cells.

Conclusions: Our results reveal the transcriptional program that could explain the enhancement of INGAP-PP-mediated physiological effects on β-cell mass and function. We identified novel lncRNAs that are induced by INGAP-PP in rat islets, some of which are selectively expressed in pancreatic β-cells and downregulated by IL1β treatment of INS-1 cells. Our results suggest a relevant function for Ri-lnc1 in β-cells. These findings are expected to provide the basis for a deeper understanding of islet translational results from rodents to humans, with the ultimate goal of designing new therapies for people with diabetes.

Keywords: INGAP; Ri-lnc1; beta cell (β-cell); islet; long noncoding RNA (IncRNA); pancreas; rat; regeneration.

MeSH terms

  • Animals
  • Diabetes Mellitus* / metabolism
  • Humans
  • Insulin / metabolism
  • Islets of Langerhans* / metabolism
  • Pancreatitis-Associated Proteins / genetics
  • Pancreatitis-Associated Proteins / metabolism
  • Pancreatitis-Associated Proteins / pharmacology
  • Peptides / metabolism
  • RNA, Long Noncoding* / genetics
  • RNA, Long Noncoding* / metabolism
  • Rats
  • Vitamin D / metabolism

Substances

  • RNA, Long Noncoding
  • Pancreatitis-Associated Proteins
  • Insulin
  • Peptides
  • Vitamin D

Grants and funding

JG, BM, LF and SR-S are career investigators from the Consejo Nacional de Investigaciones Científicas y Técnicas of Argentina (CONICET), CR is member of the Research Career of CICPBA. This work was supported by grants from the Agencia Nacional de Promoción Científica y Tecnológica of Argentina (PICT-2014 2814, PICT-2017 2071) to SR-S, BM and LF. AR, AH and MA are supported by PhD fellowships from the CONICET.