Diabetes Mellitus Type II. Beta Cell Failure in DM T2 signaling pathways implicated in β -cell failure.

Slides:



Advertisements
Similar presentations
500X 1000X Name the organ from which these slides were prepared.
Advertisements

4.A.3 Cell Specialization Interactions between external stimuli and regulated gene expression result in specialization of cells, tissues and organs.
Diabetes and Aging MCB 135K Laura Epstein 4/14/06.
PANCREAS AND DIABETES Valerija Vrhovnik Mentor: A. Žmegač Horvat.
The Endocrine System Anatomy and Physiology Endocrine System Endocrine organs secrete hormones directly into body fluids (blood) Hormones are chemical.
Jovonne Carr Cheyenne Hockenhull
BC21D Carbohydrate Metabolism Rachael Irving Biochemistry.
Caloric Restriction and Resveratrol Reverse a High-fat Diet Induced Diabetes and Improve Islet β cell Dysfunction in Mice Jiaoyue Zhang Ph.D Endocrinology.
The regulation of blood glucose Starter:  Write down as many things as you can think about that affect your blood sugar levels 04/10/2015.
A and P II Glucose Metabolism. 120 grams of glucose / day = 480 calories.
Regulation of insulin levels Starter: what do each of the following cells produce and are they part of the endocrine or exocrine system; –α cells –β cells.
Seong-Eui Hong. Background
Responses of Cells to Environmental Influences By Kari Edge.
Diabetes mellitus. Normal endocrine pancreas 1 million microscopic clusters of cells 1 million microscopic clusters of cells Β,α,δ,PP cells Β,α,δ,PP cells.
Downregulation of Wnt/ β -catenin signaling causes degeneration of hippocampal neurons in vivo April 1, 2011.
Endocrine Physiology The Endocrine Pancreas. A triangular gland, which has both exocrine and endocrine cells, located behind the stomach Strategic location.
Liver to pancreas transdifferentiation by pancreatic transcription factors Shiraz Gefen-Halevi Ph.D Proposal Dr. Sarah Ferber Prof. Jacob Shoham.
Autoimmunity and Type I Diabetes CCMD 793A: Fundamental Integrated SystemsFALL, 2006 James M. Sheil, Ph.D.
Endocrine Physiology The Endocrine Pancreas Dr. Khalid Al-Regaiey.
The Pancreas By Margaret Sheekey Histology of the Pancreas The pancreas is in the abdomen, just below the stomach It is to the right of the liver when.
Teratoma Formation Leads to Failure of Treatment for Type I Diabetes Using Embryonic Stem Cell-Derived Insulin-Producing Cells  Takahisa Fujikawa, Seh-Hoon.
The effects of β-cell specific knockdown of FoxO1 during diet-induced obesity Oluwabukola A. Ajasa, Jennifer Fiori O’Connell, Michael Rouse, Adria Summers,
From: MicroRNA let-7 Regulates 3T3-L1 Adipogenesis
Volume 119, Issue 5, Pages (November 2000)
Peter L. Lee, Yuefeng Tang, Huawei Li, David A. Guertin 
Figure 1 Body weight of control and BPA-treated mothers after delivery
Volume 90, Issue 1, Pages (July 2016)
Nithya Krishnamurthy, Razelle Kurzrock  Cancer Treatment Reviews 
Dapper-1 Induces Myocardial Remodeling Through Activation of Canonical Wnt Signaling in CardiomyocytesNovelty and Significance by Marco Hagenmueller, Johannes.
Volume 12, Issue 2, Pages (February 2013)
The gene encoding TP53INP1 is expressed in pancreatic endocrine cells A, B(A, B) Immunocytofluorescent staining of TP53INP1 (red) and insulin (green) in.
Volume 138, Issue 5, Pages e1 (May 2010)
Volume 117, Issue 6, Pages (December 1999)
Volume 90, Issue 1, Pages (July 2016)
David A. Cano, Shigeki Sekine, Matthias Hebrok  Gastroenterology 
Corona cell RNA sequencing from individual oocytes revealed transcripts and pathways linked to euploid oocyte competence and live birth  Jason C. Parks,
Volume 137, Issue 6, Pages (December 2009)
Volume 119, Issue 5, Pages (November 2000)
Volume 128, Issue 3, Pages (March 2005)
Figure 1 WNT signalling pathways
Volume 125, Issue 4, Pages (October 2003)
Volume 23, Issue 6, Pages (May 2018)
Bonnie E Lonze, David D Ginty  Neuron 
Stem Cells and Diabetes
Volume 25, Issue 11, Pages (November 2017)
The Ying and Yang of Bacterial Signaling in Necrotizing Enterocolitis
Volume 134, Issue 2, Pages e3 (February 2008)
Volume 138, Issue 5, Pages e1 (May 2010)
by Ling-juan Zhang, Christian F. Guerrero-Juarez, Tissa Hata, Sagar P
Volume 14, Issue 1, Pages (July 2011)
Communication and Homeostasis
Volume 19, Issue 11, Pages (November 2011)
Expression Pattern of Wnt Signaling Components in the Adult Intestine
Volume 17, Issue 10, Pages (October 2009)
Volume 137, Issue 5, Pages (November 2009)
Sapna Puri, Matthias Hebrok  Developmental Cell 
Volume 1, Issue 4, Pages (April 2005)
Volume 7, Issue 4, Pages (October 2016)
Evidence for Altered Wnt Signaling in Psoriatic Skin
Stem Cells and Diabetes
Volume 96, Issue 3, Pages (February 1999)
Volume 7, Issue 5, Pages (June 2014)
Increased Expression of Wnt2 and SFRP4 in Tsk Mouse Skin: Role of Wnt Signaling in Altered Dermal Fibrillin Deposition and Systemic Sclerosis  Julie Bayle,
Volume 129, Issue 2, Pages (April 2007)
Role of the amino-terminal domain of simian virus 40 early region in inducing tumors in secretin-expressing cells in transgenic mice  Christelle Ratineau,
WASH cKO mice display a normal pancreatic development.
Nat. Rev. Rheumatol. doi: /nrrheum
Glucose-stimulated insulin secretion, plasma glucagon levels, and pancreatic hormone contents. Glucose-stimulated insulin secretion, plasma glucagon levels,
Mutations in WNT10B Are Identified in Individuals with Oligodontia
Presentation transcript:

Diabetes Mellitus Type II

Beta Cell Failure in DM T2 signaling pathways implicated in β -cell failure

Wnt Signaling Pathway Controls organismal growth and differentiation

Wnt signalling Pathway and DM T2 1. Homozygous mutation of LRP5 in mice leads to defective glucose-stimulated insulin secretion from isolated islets in vitro. 2. Components of the Wnt pathway are present in the adult pancreas, and in particular multiple members of the frizzled family of Wnt receptors have been identified in the islet. 3. TCF7/L2 gene polymorphism affects B-cell function

While most studies have found little evidence that Wnt signaling is involved in endocrine differentiation or in the adult islet, there is some evidence for an effect of Wnt signaling on β-cell replication.

Main Objective To study the role of Wnt signaling in human diabetes Systematically examined components of the pathway in the pancreas of normal & DMT2 Mouse Studies

METHODS TISSUE PREPARATION 1. Paraffin-embedded from human pancreases: 5 non-diabetic & 9 DM T2 2. Freshly isolated & cultured pancreases: 3 non-diabetic (Isolated human islets) 3. Human Fetal Pancreases in gestational weeks 4. Murine Pancreaes (Balb/c or C57/bl mice) 5. NEPCs WESTERN BLOT 1. Whole-cell extracts IMMUNO-HISTOCHEMISTRY 1. Paraffin samples 2. Frozen samples

I.Objective: To determine if TCF7L2 is upregulated in islets of type II diabetic patients TCF7L2 is a protein acting as a transcription factor. Several SNPs of the gene are associated with Type 2 DM

TCF/L2TCF3 TCF7L2 is upregulated in islets of type II diabetic patients

II. Objective: To determine if Wnt2b is upregulated in type II diabetes To test the hypothesis that the induction of TCF factors, being a both effector as well as a downstream target of Wnt Signaling, is a result of a more global activation of Wnt signaling

B Catenin Wnt2b In islets Wnt 2b is upregulated in Type 2 Diabetes

III. Objective: To determine if B-catenin is upregulated in type II diabetes

B-Catenin In islets Beta-catenin in human islets of all 5 nondiabetics was markedly lower Than in the surrounding exocrine Tissue where it was strongly Expressed (figs. 2f-2h) Beta-catenin is strongly expres in the islets of all DM T2 to a Level approximately half that of the Surrounding exocrine tissue (figs 2h-2j) insulin islet insulin B-catenin insulin B-catenin DM T2 Normal Human B-cells lack Detectable B-catenin expression but it is highly upregulated in DM T2

IV. Objective: To determine if the terminal effectors (c-myc & cyclin D) are upregulated in DM T2 TCF/LEF factors activate a number of terminal effectors Of Wnt signaling (c-myc and cyclinD1)

c NORMAL DM T2 CyclinD1 In islets C-myc In islets Terminal effectors of Wnt signaling (c-myc & cyclinD1) are upregulated in human type 2 Diabetes

MOUSE MODEL V. Objective: To determine whether Wnt activation was an early or late event in diabetes pathogenesis

high-fat diet normal chow Harvested pancreases examined for c-myc expression OBESE BUT WITH NORMAL FBS 12 weeks

control High-fat control High-fat control

Figure 6: Wnt signaling in high-fat fed mice. In normal mouse pancreas, β -catenin (green in (a) and (b)) is expressed in islets as identified by somatostatin (red in (a)) and colocalizes with insulin (red in (c)). C-myc (red in (d) and (e)) was not expressed in islets of normal mice (marked by dotted lines and glucagon in green in (d)) but was induced in islets and some ducts of high-fat fed mice (islets marked by glucagon in green). C-myc expression is quantitated in (f). B -catenin somatostatin insulin Islets C-myc High-Fat Normal Glucagon Expression of the Wnt target gene c-myc is an early response to high-fat diet

Summary 1. B-catenin in human islets of all 5 nondiabetics was markedly lower than in the surrounding exocrine tissue, where it was strongly expressed. 2. Expression of the Wnt2b, B-catenin, TCF7/L2, & terminal effectors (c-myc & cyclinD1) are all upregulated in the islets in type II diabetes 3. The mouse model suggested that obesity alone may be sufficient to induce Wnt activation, which would mark it as an early event in the pathogenesis of type II diabetes.

Conclusion Type II Diabetes activates the Wnt signaling pathway specifically in the Beta cells of the islets of Langerhans

Thank You!!!

Immunohistochemical analysis of insulin (green) and anti-sFRP (red in (f) and (g)) detects sFRP on islet cells only when sFRP has been added to the culture media. sFRP exposure led to inhibition of β-catenin ((h) versus (i), quantitated in (j)) and c-myc ((k) versus (l), quantitated in (m)). Error bars = mean +/−s.e.m. ∗ P <.05. Scale bars in (f)–(m): 25 μm.

To pursue the role of Wnt activation in the islet, it would be desirable to have an in vitro model. Thus, we examined Wnt activation in isolated islets. Surprisingly, when cultured human islets were examined by Western blotting, β- catenin, which is low or absent in the islet compared with surrounding tissue in situ, was expressed at a higher level than in the nonendocrine pancreatic cells (NEPCs) [23] (Figure 4(a)).

To pursue the finding that β-cells with low- insulin expression had a pattern of catenin expression resembling that in the exocrine pancreas, pancreas sections were immunostained for the acinar marker amylase as well as insulin, revealing that low-insulin β- cells coexpressed amylase (Figures 5(f), 5(g), 5(h)). The insulin/amylase doublepositive cells expressed PDX-1 (Figure 5(i)), which in the adult pancreas is restricted to β-cells and is never expressed in mature acinar cells, indicating that the weak insulin expression was not artifactual.

To further explore whether the areas containing the insulin/amylase double-positive cells arose by alteration of preexisting β-cells or by induction of insulin expression in preexisting exocrine cells, as has been described in some β- cell regeneration models [37–39], we examined those areas for glucagon expression. Consistently, high levels of glucagon and a lack of amylase were observed in all α-cells, whether the islets exhibited high or low insulin expression (Figure 5(l)).

Figure 1. Schematic representation of WNT/TCF signalling. Secreted WNTs bind to FZD and LRP receptors, which in turn inactivate the degradation complex comprising AXIN, DVL and GSK3B. This results in non-phosphorylated b-catenin entering into the nucleus and binding to TCF7L2, thus activating a wide variety of genes. TCF7L2 could regulate several genes – tissue specifically influencing both insulin secretion and insulin sensitivity. For example, TCF7L2 regulates b-cell growth and differentiation. TCF7L2 also activates the expression of proglucagon gene, which encodes the GLP-1 (glucagons like peptide-1) and thus promotes insulin secretion. Alterations in this pathway (in TCF7L2 risk variants) could lead to reduced secretion of GLP-1 and hence defective insulin secretion. In addition, altered WNT signalling (in TCF7L2 risk variants) could be expected to influence adipose tissue growth and development and thus BMI. Increased pro-inflammatory signals (IL-6, TNF-a) and altered adiponectin from the adipocytes (through their endocrine function) might result in skeletal muscle insulin resistance. With regard to microvascular complications of diabetes, TCF7L2 may also influence mesangial cell expansion and retinal neovascularization.