Mucosal atrophy often leads to barrier failure and sepsis in starving or fasting patients despite total parenteral nutrition (TPN). Many patients who undergo massive bowel resection fail to adapt and require permanent TPN with high mortality. Current interventions to prevent mucosal atrophy or enhance adaptation have limited efficacy, perhaps because they mostly induce proliferation. We hypothesize that hyperproliferation can be maladaptive as it is often accompanied by decreased differentiation. Thus, we propose that we must learn to promote differentiation as well as proliferation to achieve an intact functional mucosa, reverse mucosal atrophy, and engender maximal intestinal adaptation. Schlafen (Slfn)-family proteins mediate cell growth, differentiation, or development in hematopoietic cells in a Slfn and cell-specific fashion. We showed that the rodent protein Slfn3 is induced during and required for intestinal epithelial differentiation in response to various stimuli. We further found that Slfn3 varies with intestinal development and mucosal atrophy and promotes differentiation in vivo and that SLFN12 is its human ortholog. We hypothesize that Slfn3/12 induction represents a fundamental and essential common pathway for enterocytic differentiation that can be specifically targeted to promote differentiation and maintain the mucosa. We further propose that Slfns act in the cytosol through Slfn P-loop domain binding to specific proteins lowering proteasomal activity, increasing Cdx2 homeobox protein levels and enhancing transcription via a Cdx2-dependent positive feedback mechanism. We will study 1) intestinal mucosa in patients fasting on TPN (atrophy) or after Roux-en-Y gastric bypass (hypertrophy), 2) rat and human intestinal epithelial cells in vitro, and 3) the effects of in vivo manipulation of Slfn3/12 levelsin Slfn3 knock-out mice after 60% massive small bowel resection and a novel rodent model of intestinal epithelial atrophy and hypertrophy to: 1) Demonstrate that Slfn3 determines enterocytic differentiation in mucosal atrophy and adaptation in rodents, 2) Show that SLFN12 is the relevant human ortholog of Slfn3, and 3) Establish the mechanism(s) by which Slfn3/SLFN12 act. This study will fill a critical knowledge gap because we will delineate a fundamental mechanism by which diverse stimuli regulate intestinal epithelial differentiation. This will facilitate interventions to maintain the mucosal barrier in starving or critically ill paients with mucosal atrophy and promote nutrition in pediatric or adult short bowel syndrome. The Slfn3 knock-out mouse will provide valuable insights not only into the convergent differentiation regulatory pathway we will study but also into normal gut development, mucosal healing and other intestinal disorders such as IBD. In addition, understanding how Slfn proteins promote differentiation will have broader implications for differentiation and development in other epithelial tissues.

Public Health Relevance

Intestinal mucosal atrophy and barrier failure are common in fasting or starving patients despite parenteral nutrition (TPN) and some patients undergoing massive small bowel resection may require permanent TPN because inadequate mucosal adaptation does not allow sufficient nutrition for survival. Therapies aimed at enhancing adaptation have shown limited success, perhaps because they have focused primarily on enhancing mucosal proliferation. In this study, we will delineate a way to promote mucosal epithelial cell differentiation in vivo, an intervention that may eventually be used to improve survival in fasting or short gut patients.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK096137-04
Application #
9135403
Study Section
Surgery, Anesthesiology and Trauma Study Section (SAT)
Program Officer
Perrin, Peter J
Project Start
2014-09-01
Project End
2018-08-31
Budget Start
2016-09-01
Budget End
2017-08-31
Support Year
4
Fiscal Year
2016
Total Cost
$284,084
Indirect Cost
$66,584
Name
University of North Dakota
Department
Type
Schools of Medicine
DUNS #
102280781
City
Grand Forks
State
ND
Country
United States
Zip Code
58202
Basson, Marc D; Wang, Qinggang; Chaturvedi, Lakshmi S et al. (2018) Schlafen 12 Interaction with SerpinB12 and Deubiquitylases Drives Human Enterocyte Differentiation. Cell Physiol Biochem 48:1274-1290
Walsh, Mary F; Hermann, Rebecca; Lee, Jun Hee et al. (2015) Schlafen 3 Mediates the Differentiating Effects of Cdx2 in Rat IEC-Cdx2L1 Enterocytes. J Invest Surg 28:202-7
Kovalenko, Pavlo L; Basson, Marc D (2014) Schlafen 12 expression modulates prostate cancer cell differentiation. J Surg Res 190:177-84
Chaturvedi, Lakshmishankar; Sun, Kelian; Walsh, Mary F et al. (2014) The P-loop region of Schlafen 3 acts within the cytosol to induce differentiation of human Caco-2 intestinal epithelial cells. Biochim Biophys Acta 1843:3029-37
Kovalenko, Pavlo L; Yuan, Lisi; Sun, Kelian et al. (2013) Regulation of epithelial differentiation in rat intestine by intraluminal delivery of an adenoviral vector or silencing RNA coding for Schlafen 3. PLoS One 8:e79745
Kovalenko, Pavlo L; Basson, Marc D (2013) The correlation between the expression of differentiation markers in rat small intestinal mucosa and the transcript levels of schlafen 3. JAMA Surg 148:1013-9
Chaturvedi, Lakshmi S; Basson, Marc D (2013) Glucagonlike peptide 2 analogue teduglutide: stimulation of proliferation but reduction of differentiation in human Caco-2 intestinal epithelial cells. JAMA Surg 148:1037-42
Walsh, Mary F; Hermann, Rebecca; Sun, Kelian et al. (2012) Schlafen 3 changes during rat intestinal maturation. Am J Surg 204:598-601