• Aucun résultat trouvé

Argonaute2 mediates compensatory expansion of the pancreatic β cell

N/A
N/A
Protected

Academic year: 2022

Partager "Argonaute2 mediates compensatory expansion of the pancreatic β cell"

Copied!
14
0
0

Texte intégral

(1)

Article

Reference

Argonaute2 mediates compensatory expansion of the pancreatic β cell

TATTIKOTA, Sudhir G, et al.

Abstract

Pancreatic β cells adapt to compensate for increased metabolic demand during insulin resistance. Although the microRNA pathway has an essential role in β cell proliferation, the extent of its contribution is unclear. Here, we report that miR-184 is silenced in the pancreatic islets of insulin-resistant mouse models and type 2 diabetic human subjects. Reduction of miR-184 promotes the expression of its target Argonaute2 (Ago2), a component of the microRNA-induced silencing complex. Moreover, restoration of miR-184 in leptin-deficient ob/ob mice decreased Ago2 and prevented compensatory β cell expansion. Loss of Ago2 during insulin resistance blocked β cell growth and relieved the regulation of miR-375-targeted genes, including the growth suppressor Cadm1. Lastly, administration of a ketogenic diet to ob/ob mice rescued insulin sensitivity and miR-184 expression and restored Ago2 and β cell mass. This study identifies the targeting of Ago2 by miR-184 as an essential component of the compensatory response to regulate proliferation according to insulin sensitivity.

TATTIKOTA, Sudhir G, et al . Argonaute2 mediates compensatory expansion of the pancreatic β cell. Cell Metabolism , 2014, vol. 19, no. 1, p. 122-34

DOI : 10.1016/j.cmet.2013.11.015 PMID : 24361012

Available at:

http://archive-ouverte.unige.ch/unige:55459

Disclaimer: layout of this document may differ from the published version.

(2)

Argonaute2 Mediates Compensatory Expansion of the Pancreatic b Cell

Sudhir G. Tattikota,1,14Thomas Rathjen,1,14Sarah J. McAnulty,1Hans-Hermann Wessels,1Ildem Akerman,5

Martijn van de Bunt,7Jean Hausser,8Jonathan L.S. Esguerra,9Anne Musahl,1Amit K. Pandey,1Xintian You,1Wei Chen,1 Pedro L. Herrera,10Paul R. Johnson,7,11,12Donal O’Carroll,13Lena Eliasson,9Mihaela Zavolan,8Anna L. Gloyn,7,11 Jorge Ferrer,5,6Ruby Shalom-Feuerstein,4Daniel Aberdam,2,3and Matthew N. Poy1,*

1Max Delbrueck Center for Molecular Medicine, 13125 Berlin, Germany

2Institut National de la Sante´ et de la Recherche Me´dicale (INSERM), UMR-S 976, 75475 Paris, France

3Universite´ Paris Diderot, Skin Research Center, 75013 Paris, France

4Department of Anatomy and Cell Biology, The Ruth and Bruce Rappaport Faculty of Medicine, The Technion-Israel Institute of Technology, 31096 Haifa, Israel

5Genomic Programming of Beta-Cells Laboratory, Institut d’Investigacions Biome`diques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain

6Department of Medicine, Imperial College London, W12 0NN London, UK

7Oxford Centre for Diabetes, Endocrinology, & Metabolism, University of Oxford, OX3 7LE Oxford, UK

8Computational and Systems Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland

9Department of Clinical Sciences, Lund University Diabetes Center, Lund University, Malmo¨ University Hospital, SE-205 02 Malmo¨, Sweden

10Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland

11NIHR Oxford Biomedical Research Centre, ORH Trust, OCDEM, Churchill Hospital, OX3 7LJ Oxford, UK

12Nuffield Department of Surgery, University of Oxford, OX3 9DU Oxford, UK

13European Molecular Biology Laboratory, 00015 Monterotondo Scalo, Italy

14These authors contributed equally to this work

*Correspondence:matthew.poy@mdc-berlin.de http://dx.doi.org/10.1016/j.cmet.2013.11.015

SUMMARY

Pancreatic

b

cells adapt to compensate for increased metabolic demand during insulin resis- tance. Although the microRNA pathway has an essential role in

b

cell proliferation, the extent of its contribution is unclear. Here, we report that

miR- 184

is silenced in the pancreatic islets of insulin- resistant mouse models and type 2 diabetic human subjects. Reduction of

miR-184

promotes the expression of its target

Argonaute2

(Ago2), a compo- nent of the microRNA-induced silencing complex.

Moreover, restoration of

miR-184

in

leptin-deficient ob/ob

mice decreased

Ago2

and prevented compen- satory

b

cell expansion. Loss of

Ago2

during insulin resistance blocked

b

cell growth and relieved the regulation of

miR-375-targeted genes, including the

growth suppressor

Cadm1. Lastly, administration

of a ketogenic diet to

ob/ob

mice rescued insulin sensitivity and

miR-184

expression and restored

Ago2

and

b

cell mass. This study identifies the targeting of

Ago2

by

miR-184

as an essential compo- nent of the compensatory response to regulate pro- liferation according to insulin sensitivity.

INTRODUCTION

Adaptation to environmental stress is a fundamental cellular pro- cess that promotes the maintenance of the physiologic steady

state (Spriggs et al., 2010). Stress responses have been shown to induce numerous changes, such as activation of gene expres- sion programs, which have evolved to allow for the cell to pro- mote its own survival (Ku¨ltz, 2005; Ebert and Sharp, 2012). For example, in response to insulin resistance, the pancreatic b cell undertakes measures to proliferate and increase its output of secreted insulin. A coordinated increase in bothbcell mass and secretory function constitutes the compensatory response to maintain normoglycemia (Muoio and Newgard, 2008).

Although the underlying mechanisms directing these processes are still not completely understood, several studies have illus- trated a role for metabolic changes in catalyzingbcell expansion (Steil et al., 2001). Furthermore, cellular pathways enabling theb cells to proliferate and adapt to increases in metabolic load may act by ultimately promoting signaling cascades essential to increasing both secretion and islet mass (Rhodes, 2005).

Recent evidence has shown the microRNA (miRNA) pathway as an important regulator of gene expression in response to metabolic stress (Leung and Sharp, 2010). Central to this mechanism are the Argonaute (Ago) proteins, which mediate this pathway by facilitating the interaction between miRNAs and their target mRNAs (Ho¨ck and Meister, 2008; Bartel, 2009). In addition, Ago proteins have been shown to accumulate in stress granules upon exposure to oxidative stress; however, their role in this compartment is not understood (Leung et al., 2006). Although loss of Argonaute2 (Ago2) expression in the MIN6bcell line model resulted in enhanced secretion, its role in the stress response of the b cell has not been described (Tattikota et al., 2013).

We have previously shown that loss ofmiR-375expression, among the most abundant miRNA in the pancreatic islet, inhibited the compensatorybcell proliferation inleptin-deficient Open access under CC BY license.

(3)

ob/obmice and resulted in severe hyperglycemia and diabetes (Poy et al., 2009). The absence of any dramatic effect on the development or specification of the different cell populations in themiR-375knockout mouse may indicate a larger role for this miRNA in stress responses (Mendell and Olson, 2012). Further- more, these observations suggest that many of the targets of miR-375 are also relevant to the adaptive response of theb cell and likely play a role in proliferation during metabolic stress.

Although extensive sequencing efforts have identified2,000 mature miRNA sequences in human tissues, relatively little is understood regarding how small RNAs coordinately function in these cellular processes (Kozomara and Griffiths-Jones, 2011).

Here, we show thatmiR-184is silenced during insulin resis- tance to promote the expression of Ago2 in the pancreaticb cell. Deletion ofAgo2inob/obmice reduced compensatory pro- liferation of this cell type, thereby underlining an integral role for the miRNA pathway in this process. Moreover, we observed that Ago2 mediates the function ofmiR-375in regulating the growth suppressor Cadm1. Taken together, our results show that several components of the miRNA pathway contribute in a

concerted effort to facilitate proliferation of the bcell to meet metabolic demand during insulin resistance.

RESULTS

Silencing ofmiR-184in the Pancreaticb-Cell Promotes Its Target Argonaute2

In light of the essential role ofmiR-375in adaptive growth of the pancreaticbcell, we first sought to identify the additional com- ponents of the miRNA pathway that coordinately mediate this mechanism. We performed small RNA sequencing on total RNA from islets of 12-week-oldob/obmice (Table S1available online). Consistent with results byZhao et al. (2009), expression ofmiR-184was the most reduced miRNA identified (Figure 1A;

Table S1). We then measuredmiR-184 in the islets of ob/ob mice from age 4–16 weeks and observed the decrease in expression starting at 8 weeks of age with the onset of resistance (Figures 1B,S1A, and S1B). Similarly, the pri-miR-184transcript in the islets ofob/obmice by quantitative real-time PCR was also silenced, indicating that this miRNA is regulated on a 0

0.4 0.8 1.2 1.6

4 8 12 16

age (weeks) miR-184(AU) ob/ob

WT

** *** ***

0 0.4 0.8 1.2 1.6

4 8 12 16

age (weeks) miR-184(AU) ob/ob

WT

** *** ***

0 0.4 0.8 1.2

WTob/ob

pri-miR-184(AU)

**

0 0.4 0.8 1.2

WTob/ob

pri-miR-184(AU)

**

B C

J K

H

**

I

0.0 1.5

mAgo2 norm. luc. (Rr/Pp)

mAgo2 MUT mAgo2

MUT

**

hAgo2 hAgo2 hAgo2

MUT hAgo2

MUT 1.0

0.5

0.0 1.5

mAgo2 norm. luc. (Rr/Pp)

hAgo2 1.0

0.5

+ ctrl-mimic + MUT-184 n.s. n.s.

0.0 1.5

mAgo2 norm. luc. (Rr/Pp)

hAgo2 1.0

0.5

+ ctrl-mimic + MUT-184 + ctrl-mimic + MUT-184 n.s. n.s.

miR-184 miR-375

1 10 100 10000 1000000

110100100001000000

WT islet (reads)

ob/obislet (reads)

A

miR-184 miR-375

1 10 100 10000 1000000

110100100001000000

WT islet (reads)

ob/obislet (reads)

0 0.4 0.8 1.2

WTdb/db

miR-184(AU)

0

***

0.4 0.8 1.2

WTdb/db

miR-184(AU)

***

D F Chow G

HFD

0.0 1.0 2.0

8 16 20

* ***

weeks on HFD

miR-184(AU)

Chow HFD Chow HFD

0.0 1.0 2.0

8 16 20

* ***

weeks on HFD

miR-184(AU)

Chow HFD Chow HFD

0.0 1.0 2.0

20 weeks on

HFD

miR-375(AU)

*

WT ob/ob WT ob/ob

0.0 1.0 2.0

miR-375(AU)

*

GFP- E

GFP+

miR-184(AU)

0 5 10 15

GFP- GFP+

GFP- GFP+

miR-184(AU)

0 5 10 15

0 5 10 15

ctrl-mimic 184-mimic

Ago2 γ-tubulin

50 pM 200 pM 200 pM

ctrl-mimic 184-mimic

Ago2 γ-tubulin

50 pM 200 pM 200 pM

miR-184(AU) (Norm. to Control Islets) 0.0 0.5 1.0 1.5

**

Non T2D miR-184(AU) (Norm. to Control Islets)

0.0 0.5 1.0 1.5

**

Non T2D

L M

R = - 0.334

0 1 2 3

0 1 2

Ago2(AU)

miR-184 (AU) T2D Non

p< 0.05 R = - 0.334

0 1 2 3

0 1 2

Ago2(AU)

miR-184 (AU) T2D Non

p< 0.05

0.0 1.0 2.0 3.0

Non T2D Ago2mRNA (Norm. to Control Islets)

0.0 1.0 2.0 3.0

Non T2D Ago2mRNA (Norm. to Control Islets)

0 1 2 3

Non T2D miR-184(AU) (Norm. to Control Islets)

**

0 1 2 3

Non T2D miR-184(AU) (Norm. to Control Islets)

0 1 2 3

Non T2D miR-184(AU) (Norm. to Control Islets)

**

50 200 ctrl-mimic 184-mimic

Ago2mRNA (AU)

** **

0.0 0.5 1.0 1.5

pM pM50 200 ctrl-mimic 184-mimic ctrl-mimic 184-mimic

Ago2mRNA (AU)

** **

0.0 0.5 1.0 1.5

0.0 0.5 1.0 1.5

pM pM Ago2 protein exp. (normalized)

0.0 0.5 1.0 1.5

ctrl 50 200

**

*

pM pM Ago2 protein exp. (normalized)

0.0 0.5 1.0 1.5

ctrl 50 200

**

*

pM pM ctrl 50 200

**

*

pM pM +ctrl-mimic

+184-mimic +ctrl-mimic +184-mimic

Figure 1. miR-184 Is Silenced during Insulin Resistance and Directly Targets Argonaute2

(A) Comparison of small RNA sequencing analysis from total RNA from islets of 12-week-oldob/ob and wild-type (WT) littermates.

(B) Quantitative real-time PCR analysis ofmiR-184 in islets ofob/oband WT mice from 4–16 weeks of age (n = 3–5).

(C) Quantitative real-time PCR analysis of pri-miR- 184in islets ofob/obmice and WT littermates at 16 weeks of age (n = 3–5).

(D) Quantitative real-time PCR analysis ofmiR-184 in FACS-sortedbcells from 16-week-old mouse insulinpromoter-GFP mice (n = 6).

(E) Quantitative real-time PCR analysis ofmiR-184 in islets ofdb/dbmice and WT littermates at age 12 weeks (n = 4).

(F) Quantitative real-time PCR analysis ofmiR-184 in islets of C57BL/6 mice on high-fat diet (HFD) or chow diet.

(G) Quantitative real-time PCR analysis ofmiR-375 in islets of C57BL/6 mice on HFD or chow diet and in islets of ob/ob mice and littermates at age 12 weeks (n = 6).

(H and I) Luciferase assays in MIN6 cells testing direct targeting of mouse and humanAgo2genes bymiR-184(184-mimic) or mutant (MUT-184).

(J and K) Western blot and quantitative real-time PCR analysis ofAgo2after transfection ofmiR- 184-mimic and scrambled control.

(L) Quantitative real-time PCR analysis ofmiR-184 and Ago2in islets from nondiabetic (Non) and type-2 diabetic human subjects (T2D) after normalization to RNU6bandTBP, respectively, expressed as fraction of control islets. p values represent a Mann-Whitney significance test (p = 0.009 formiR-184).

(M) Correlation of quantitative real-time PCR analysis from (L) ofAgo2andmiR-184in individual T2D (n = 12) and nondiabetic (n = 15) human subjects. Results presented as mean ± SEM. *p <

0.05; **p < 0.01; ***p < 0.001.

See alsoFigure S1andTables S1andS2.

(4)

transcriptional level (Figure 1C). As recently described,miR-184 is enriched in pancreaticbcells as shown by quantitative real- time PCR from fluorescence-activated cell sorting (FACS)- sorted, GFP-positivebcells (MIP-GFP) (Figure 1D) (Hara et al., 2003; van de Bunt et al., 2013). We also observed a similar loss of expression of maturemiR-184in the islets of 12-week- oldleptin receptor-deficientdb/dbmice and in mice on a high- fat diet (HFD; 60% calories from fat), all of which showed that this observation is not limited to one mouse model of obesity and insulin resistance (Figures 1E and 1F). In contrast tomiR- 184,miR-375 was modestly increased in HFD-fed animals as previously observed in ob/ob mice (Figure 1G) (Poy et al., 2009). In addition, the suppression ofmiR-184was not observed in the eye ofob/obmice, the highest site at which expression has been measured (Figures S1C and S1D).

In order to identify direct targets ofmiR-184, we induced its expression in the MIN6 line with doxycycline after transfection with plasmids expressing the rtTA transactivator andmiR-184 under control of the operator sequence of theEscherichia coli tetracycline-resistance operon (184-tetO). Illumina WG6v2 arrays measured changes in gene expression after 16 hr of treat- ment with doxycycline in triplicate (Figure S1E). Several compu- tationally predicted genes formiR-184were downregulated in both the array and luciferase-based experiments, including Ago2(Figure 1H;Figures S1F–S1H) (Friedman et al., 2009). We cloned a portion of the mouse 30UTR ofAgo2(912 nt) and the complete human UTR ofAgo2(899 nt) into a luciferase reporter construct and observed decreased activity in the presence of the miR-184mimic. In addition, mutating six nucleotides in the bind- ing site within the mouse or human UTR (at position 152) from UCCGUCC to CGGGCGG abolished the inhibitory effect of miR-184 (Figure 1H). Conversely, transfecting a mutant miR- 184mimic had no effect on reporter activity from constructs bearing either the mouse or humanAgo2 30 UTR sequences (Figure 1I). Western blotting after overexpression of miR-184 resulted in an approximately 60% decrease in Ago2 protein levels, whereas quantitative real-time PCR revealed a 40%

decrease inAgo2mRNA in MIN6 cells (Figures 1J and 1K).

To address the relevance of this microRNA:target interaction in the islets of human subjects, we measured the expression of miR-184andAgo2in the pancreatic islets from 15 nondiabetic and 12 type 2 diabetic (T2D) donors.miR-184was significantly decreased in T2D islets, indicating that the expression of this specific miRNA is linked to changes in metabolic status in both mice and humans (Figure 1L; Table S2). Whereas levels of Ago2were not statistically significantly increased in T2D donor islets compared to the nondiabetic cohort, the inverse correla- tion between Ago2 and miR-184 expression was significant across the entire cohort (Figure 1M).

Argonaute2 Regulatesb-Cell Proliferation

We next measured Ago2 expression in the islets of insulin-resis- tantob/obmice and observed increased levels at 8 weeks of age, whereas Ago1 was not altered (Figure 2A). Similarly, Ago2 levels were increased in islets isolated from 12-week-olddb/

dbmice and C57BL/6 mice on a HFD for 16 weeks (Figures 2B). As shown in other tissues,Ago2is the most abundant of the Ago family in GFP-positivebcells as measured by quantita- tive real-time PCR (Figures 2C andS2A) (Wang et al., 2012). To

understand the contribution ofAgo2to growth and function of the bcell, we generated transgenic mice bearing the mouse Ago2 cDNA under the regulatory control of a tetracycline- responsive promoter element and crossed them with mice expressing the reverse tetracycline-controlled transactivator (rtTA) protein under the control of the ratinsulin2(Ins-rtTA) pro- moter as described (dox-Ago2) (Nir et al., 2007). Administration of doxycycline via drinking water for 30 days to four different lines (14, 30, 61, and 69) resulted in1.5- to 25-fold increase of Ago2 protein levels in isolated islets (Figures 2D andS2B).

Characterization of all lines showed no changes in body weight, random glucose, or insulin levels. After a glucose challenge, dox-Ago2mice (line 30) exhibited transiently elevated glucose levels compared to littermates; however, no difference was observed in insulin sensitivity (Figures S2C and S2D). Moreover, acute-phase plasma insulin levels were diminished after glucose challenge, indicating reduced insulin release (Figure S2E).

Morphometric analysis in dox-Ago2 (line 30) animals showed increased bcell mass in addition to increased insulin-positive cell number and BrdU incorporation compared to littermates (Figures 2E–2H).

We then established the conditional deletion in b cells by crossing mice bearing the floxed allele of theAgo2gene with animals expressing Cre-recombinase under control of the rat insulin promoter (bAgo2KO) (O’Carroll et al., 2007; Herrera, 2000). Quantitative RT-PCR and western blotting analysis confirmed an efficient reduction in Ago2 expression in islets from bAgo2KO mice compared to littermate controls (Figures 2I and 2J). In contrast, Ago1 was upregulated in islets isolated frombAgo2KO mice, suggesting a degree of compensation by this family member on the protein level (Figure 2J). Both random-fed and fasted glucose and insulin levels were un- changed; however,bAgo2KO mice exhibited improved glucose tolerance without any alteration in insulin sensitivity at 10 weeks of age compared to littermates (Figures S2F and S2G). As previ- ously observed in the MIN6 model, loss of Ago2 expression in vivo resulted in enhanced secretion of insulin after glucose challenge (Figure S2H) (Tattikota et al., 2013). In contrast to dox-Ago2 mice, we observed decreased b cell mass and number, BrdU incorporation, and pancreatic insulin content in bAgo2KO animals without any change in islet architecture, a cell number, or total pancreatic mass (Figures 2K–2N,S2I, and S2J). Moreover, no change was detected in TUNEL-positiveb cells, indicating cell death was not a primary mechanism for the effect onbcell mass (Figure 2O). Lastly, we measured an increase in both bcell size and the total number of granules (Nv, measured as LDCV volume density), whereas the number of docked granules remained unchanged (Ns, LDCV surface density) inbAgo2KO mice compared to their littermates using electron micrographs (Figure S2K).

Loss ofmiR-184Expression Promotesb-Cell Proliferation

To determine the functional role ofmiR-184in thebcell, we char- acterized the constitutivemiR-184-knockout mouse (184KO) for changes in b cell proliferation, insulin release, and glucose homeostasis. The loss of miR-184 expression resulted in decreased fasted glucose levels and increased fasted plasma insulin levels (Figures 3A–3C). Consistent with loss ofmiR-184

(5)

expression during insulin resistance, morphometric analysis of pancreatic sections from 184KO mice showed an increase inb cell mass and number and BrdU incorporation rate (Figures 3D–3F). Moreover, in line with islet expression analysis in insulin-resistant models, Ago2 expression was increased in

miR-184-deficient islets (Figures 3G and 3H). Knockout mice exhibited transiently elevated insulin release and improved toler- ance after glucose challenge, indicating additional targets, such as Slc25a22, an established regulator of insulin release, may mediate the effect ofmiR-184on secretion (Figures S1H,S3A,

A B

Ago 1

Ago 2

Ago 3

Ago 4 relativeAgo expression (AU)

0.0 2.0 4.0 6.0

Ago 1

Ago 2

Ago 3

Ago 4 relativeAgo expression (AU)

0.0 2.0 4.0 6.0

0.0 2.0 4.0 6.0

D

H

F

dox-Ago2 WT

insulin brdU

dox-Ago2 WT

insulin brdU

K L

0 10 20

WT KO gcg+ cell number (cells/mm2pancreas)

0 20 40

WT KO

*

ins+ cell number (cells/mm2pancreas) 0 20 40

WT KO

*

ins+ cell number (cells/mm2pancreas)

WT 0.0 2.0 4.0 6.0

BrdU+ cells / insulin+ cells (%)

**

Ago2 dox- Ago2 dox- E

1.01

Ago2 24.5

β-actin

WT Ago2 dox-

fold

1.01

Ago2 24.5

β-actin

WT Ago2 dox- Ago2 dox-

fold

Ago2 dox- Ago2 dox-

C

G

0.0 1.0 2.0

Ago 1

Ago 2 relative expression (AU)

***

WTβAgo2KO

Ago 3

Ago 4 0.0

1.0 2.0

Ago 1

Ago 2 relative expression (AU)

***

WTβAgo2KO WTβAgo2KO

Ago 3

Ago 4 I

WT

Ago2 0.03

γ-tubulin 1.22

βAgo2KO fold

Ago1 1.80

WT

Ago2 0.03

γ-tubulin 1.22

βAgo2KO fold

Ago1 1.80

J

M WT ob/ob fold

Ago2 1.39

**

γ-tubulin 1.08

Ago1 0.77

WT ob/ob fold

Ago2 1.39

**

γ-tubulin 1.08

Ago1 0.77

WT ob/ob fold

Ago2 1.39

**

γ-tubulin 1.08

Ago1 0.77

Ago2 dox- 0

30 60

WT

*

ins+ cell number (cells/mm2pancreas)

Ago2

dox- 0

10 20

gcg+ cell number (cells/mm2pancreas) n.s.

WT Ago2

dox- Ago2 dox- 0

30 60

WT

*

ins+ cell number (cells/mm2pancreas)

Ago2

dox- 0

10 20

gcg+ cell number (cells/mm2pancreas) n.s.

WT 0

30 60

WT

*

ins+ cell number (cells/mm2pancreas)

Ago2 dox- Ago2

dox- 0

10 20

gcg+ cell number (cells/mm2pancreas) n.s.

WT

WT KO

β-cell mass (mg)

0 1 2

3

*

WT KO

β-cell mass (mg)

0 1 2 3

0 1 2

3

*

WT KO TUNEL+ cells/ insulin+ cells (%)

0.0 0.5 1.0 1.5

n.s.

O

0 4 8 12 16 20

WT KO pancreatic mass (mg/g BW)

0 4 8 12 16 20

WT KO

pancreatic mass (mg/g BW) n.s.

WT β-cell mass (mg)

*

0 1 2 3

WT β-cell mass (mg)

*

0 1 2 3

N

WT BrdU+ cells / insulin+ cells (%)

KO

*

0 1 2 3 4

WT BrdU+ cells / insulin+ cells (%)

KO

*

0 1 2 3 4 0.0 1.0 2.0

*

Ago2mRNA(AU) Chow

HFD

Ago2mRNA(AU)

*

0 1 2 3

WTdb/

db 0.0

1.0 2.0

0.0 1.0 2.0

*

Ago2mRNA(AU) Chow

HFD Chow HFD

Ago2mRNA(AU)

*

0 1 2 3

0 WT 1 2 3

WTdb/

db

Figure 2. Argonaute2Is Increased during Insulin Resistance and Promotesb-Cell Proliferation (A) Western blot analysis of Ago1 and Ago2 from islets of 8-week-oldob/obmice and wild-type (WT) littermates.

(B) Quantitative real-time PCR analysis ofAgo2in islets after 16 weeks HFD and from 12-week-olddb/dbmice (n = 3–6).

(C) Quantitative real-time PCR analysis ofArgonautegenes in FACS-sortedbcells from 12-week-old MIP-GFP mice (n = 3).

(D) Western blot analysis of Ago2 from islets of 10-week-olddox-Ago2mice (line 30) and WT littermates.

(E)bcell mass analysis of 10-week-olddox-Ago2mice (line 30) and WT (n = 3).

(F) Morphometric analysis of insulin+ and glucagon+ cells indox-Ago2and WT at age 10 weeks (n = 5–6).

(G and H) Ratio of BrdU (red) and insulin+ cells (green) in 10-week-olddox-Ago2mice and WT littermates (n = 4) after immunostaining. Scale bars = 30mm.

(I) Quantitative real-time PCR analysis ofArgonautefamily members in islets of 10-week-oldbAgo2KO mice and littermates (n = 4).

(J) Western blot analysis of Ago2 and Ago1 from islets of 10-week-oldbAgo2KO mice and WT.

(K)bcell mass analysis of 10-week-oldbAgo2KO and WT mice (n = 3).

(L) Morphometric analysis of insulin+ and glucagon+ cells inbAgo2KO and littermates at 10 weeks of age (n = 5–6).

(M) Ratio of BrdU+ and insulin+ cells inbAgo2KO and littermates at 10 weeks of age (n = 5–6).

(N) Pancreatic mass to total body mass ratio ofbAgo2KO and littermates at 10 weeks of age (n = 5).

(O) Ratio of TUNEL+ and insulin+ cells inbAgo2KO and littermates (n = 3). Results presented as mean ± SEM. *p < 0.05; **p < 0.01.

See alsoFigure S2.

(6)

and S3B) (Casimir et al., 2009). Lastly, insulin sensitivity was unchanged in 184KO mice, indicating that loss of this miRNA in thebcell does not contribute to insulin resistance (Figure 3I).

Restored Expression ofmiR-184inob/obMice Inhibits Ago2 andb-Cell Proliferation

Two gain-of-function models formiR-184were then generated in order to restore its expression in thebcell and validate the regu-

lation ofAgo2by this miRNA during insulin resistance. Similar to dox-Ago2mice, Ins2-rtTA mice were crossed to newly gener- ated mice bearing the mousemiR-184 precursor under regu- latory control of a tetracycline-responsive promoter element (dox-184). Administration of doxycycline for 15 days via drinking water (1 mg/ml) resulted in 100-fold increase in expression (Figure 3J). No change was observed in body weight; however, dox-184animals exhibited decreased plasma insulin levels, b

J K N

0 50 100 150

miR-184/ U6 (AU)

WTdox- 184 0 50 100 150

miR-184/ U6 (AU)

WTdox- 184 O

L

*

BrdU+ cells / insulin+ cells (%) 0.0 1.0 2.0 3.0

WTdox- 184

*

BrdU+ cells / insulin+ cells (%) 0.0 1.0 2.0 3.0

WTdox- 184 BrdU+ cells / insulin+ cells (%)

0.0 1.0 2.0 3.0

WTdox- 184 insulin + cells/ mm2 pancreas

0 40 80

WTdox- 184

*

insulin + cells/ mm2 pancreas 0 40 80

WTdox- 184

*

gcg+ cells/ mm2 pancreas 0 20 40

WTdox- 184 n.s.

gcg+ cells/ mm2 pancreas 0 20 40

WTdox- 184 n.s.

Ago2 γ-tubulin

ctrl- dox

dox- 184

ctrl- dox/ob

dox- 184ob Ago2

γ-tubulin

ctrl- dox

dox- 184

ctrl- dox/ob

dox- 184ob

0.0 1.0 2.0

ctrl- dox

dox- 184

dox- 184

ob ctrl- dox/

ob Ago2 expression (normalized)

1.0 0.6

1.4 0.7

0.0 1.0 2.0

ctrl- dox

dox- 184

dox- 184

ob ctrl- dox/

ob Ago2 expression (normalized)

1.0 0.6

1.4 0.7

dox- 184 ob ctrl- dox/

ob

miR-184/ U6 (AU)

0 100 200 300 400

ctrl- dox

dox- 184 ob ctrl- dox/

ob

miR-184/ U6 (AU)

0 100 200 300 400

ctrl- dox 0.0

0.5 1.0 1.5 2.0

WTdox- 184

plasma insulin (ng/ml)

*

0.0 0.5 1.0 1.5 2.0

WTdox- 184

plasma insulin (ng/ml)

*

M

WTdox- 184

*

β-cell mass (mg)

0 1 2 3

WTdox- 184

*

β-cell mass (mg)

0 1 2 3

0 1 2 3

A B C D

G F

0 50 100 150

blood glucose (mg/dL)

WT 184KO

0 15 60 120

time post inj. (min)30

*

0 50 100 150

blood glucose (mg/dL)

WT 184KO WT 184KO

0 15 60 120

time post inj. (min)30

*

H

0 20 40

WT KO

*

ins+ cell number (cells/mm2pancreas)

WT KO gcg+ cell number (cells/mm2pancreas)

0 10 20

0 20 40

WT KO

*

ins+ cell number (cells/mm2pancreas)

WT KO gcg+ cell number (cells/mm2pancreas)

0 10 20

WT KO gcg+ cell number (cells/mm2pancreas)

0 10 20

0.0 0.5 1.0

WT KO

*

β-cell mass (mg)

0.0 0.5 1.0

WT KO

*

β-cell mass (mg)

WT 184KO

miR- 184

miR- 375 relative expression (AU)

***

0.0 0.5 1.0 1.5

WT 184KO WT 184KO

miR- 184

miR- 375 relative expression (AU)

***

0.0 0.5 1.0 1.5

0.0 0.5 1.0 1.5

WT KO

*

Ago2mRNA (AU) 0.0 0.5 1.0 1.5

0.0 0.5 1.0 1.5

WT KO

*

Ago2mRNA (AU) WT

184KO

blood glucose (mg/dL) 0 50 100

150

**

6-hr fast rand.

fed WT 184KO WT 184KO

blood glucose (mg/dL) 0 50 100

150

**

6-hr fast rand.

fed 6-hr fast rand.

fed

I

1.68 Slc25a22

γ-tubulin

WT 184KO Ago2

fold 1.87

1.02 1.68 Slc25a22

γ-tubulin

WT 184KO Ago2

fold 1.87

1.02

E

plasma insulin (ng/mL)

*

WT KO 0.0 0.5 1.0

plasma insulin (ng/mL)

*

WT KO 0.0 0.5 1.0

BrdU+ cells / insulin+ cells (%)

WT KO

*

0.0 1.0 2.0

BrdU+ cells / insulin+ cells (%)

WT KO

*

0.0 1.0 2.0

*

0.0 1.0 2.0

P

0.0 0.5 1.0 1.5 2.0

TUNEL+ cells/ insulin+ cells (%)

WTdox- 184 n.s.

0.0 0.5 1.0 1.5 2.0

TUNEL+ cells/ insulin+ cells (%)

WTdox- 184 n.s.

Figure 3. miR-184RegulatesAgo2and Pancreaticb-Cell Proliferation In Vivo

(A) Quantitative real-time PCR analysis ofmiR-184andmiR-375in islets of 10-week-old 184KO mice and wild-type (WT) littermates (n = 4).

(B) Blood glucose of 10-week-old 184KO mice and littermates (n = 4).

(C) Fasted plasma insulin levels of 10-week-old 184KO mice and littermates (n = 4).

(D)bcell mass analysis of 10-week-old 184KO and WT mice (n = 4).

(E) Morphometric analysis of insulin+ and glucagon+ cells in 184KO and WT at 10 weeks of age (n = 3).

(F) Ratio of BrdU (red) and insulin+ cells (green) in 12-week-old 184KO mice and WT (n = 3).

(G) Western blot analysis of Ago2, Slc25a22, andg-tubulin from islets of 184KO mice and WT.

(H) Quantitative real-time PCR analysis ofAgo2in islets of 10-week-old 184KO mice and littermates (n = 4–5).

(I) Blood glucose levels during an ITT on 10-week-old 184KO mice and WT (n = 4–5).

(J) Quantitative real-time PCR analysis ofmiR-184in islets of 12-week-olddox-184mice and WT mice after 15 days on doxycycline (n = 4).

(K) Plasma insulin levels of 10-week-olddox-184mice and WT after 15 days on doxycycline (n = 4).

(L)bcell mass analysis of 10-week-olddox-184mice and WT after 15 days on doxycycline (n = 3).

(M) Morphometric analysis of insulin+ and glucagon+ cells in 10-week-olddox-184mice and WT (n = 4).

(N) Ratio of BrdU and insulin+ cells indox-184mice and WT (n = 4).

(O) Ratio of TUNEL+ and insulin+ cells indox-184mice and WT (n = 3).

(P) Western blot analysis of Ago2 andg-tubulin after ex vivo treatment of doxycycline on the islets ofdox-184anddox-184obmice compared to islets from respective control lean orob/oblittermates. Densitometry is normalized tog-tubulin expression ofctrl-doxislets. Quantitative real-time PCR analysis ofmiR-184 indox-184obmice andob/oblittermates. Results presented as mean ± SEM. *p < 0.05; **p < 0.01.

See alsoFigure S3.

(7)

cell mass and number, and BrdU incorporation (Figures 3K–3N andS3C). The number of apoptotic cells was unchanged, indi- cating the hyperglycemia (>250 mg/dl) and glucose intolerance indox-184mice were due to a decrease in b cell mass and rate of proliferation (Figures 3O andS3D–S3F). Isolated islets fromdox-184mice treated with doxycycline for 48 hr ex vivo ex- hibited decreasedAgo2expression (Figure 3P). Moreover, we crosseddox-184mice onto theob/obbackground (dox-184ob) and observed a 50% reduction in Ago2 expression after doxycy- cline treatment and restoring expression ofmiR-184(Figure 3P).

Furthermore, whereas islet levels ofmiR-375and Ago1 were un- changed, random blood glucose levels increased and plasma insulin levels were significantly reduced indox-184obmice (Fig- ures S3G–S3J). An additional transgenic model of constitutive overexpression was developed to further confirm the role of miR-184inbcell growth and function. We focused on three inde- pendent mouse lines (Tg-96,Tg-04, andTg-32), which showed

by Southern analysis varying numbers of the transgene contain- ing the miR-184 precursor sequence under control of the rat insulinpromoter (Figure S3K). All lines exhibited a mild decrease in body weight, but the highest overexpression of miR-184 (100-fold in Tg-04and 32) resulted in severe hyperglycemia and reduced systemic insulin levels, whereas a 5-fold increase (Tg-96) induced glucose intolerance and impaired insulin release (Figures S3L–S3Q). Similar todox-184animals,Tg-04andTg-32 animals exhibited hyperglycemia and reduced circulating insulin due to loss ofbcell mass and pancreatic insulin content (Figures 4A–4G andS4A–S4G). CrossingTg-04animals onto theob/ob background (04ob) resulted in sustained expression of miR- 184and did not compromise the most abundant miRNAs, such asmiR-375(Figures 4A and 4B). As indox-184obmice,04ob mice exhibited severe hyperglycemia as a result of diminished insulin levels andbcell mass and ultimately contributed to weight loss (Figures 4C–4I). Lastly, restoring expression ofmiR-184inb E

0.0 1.0

WT 04

ob

miR-375(AU)

ob/

ob ob/

ob Tg-

04 Tg-

04 2.0

A D

G

C

0 200 400 600 800

WTTg- 04

ob/

ob 04 ob blood glucose (mg/dL)

B

***

***

plasma insulin (ng/mL) 0 10 40 60

*

WT Tg- 04

ob/

ob 04 ob

***

plasma insulin (ng/mL) 0 10 40 60

*

WT Tg- 04

ob/

ob 04 ob

insulin content (μg /g pancreas) 04ob

***

ob/ob ob/ob Tg-04

Tg-04 WT

insulin glucagon

I

0.0 1.0 2.0

WTob/

ob 04 ob

Ago2mRNA(AU)

*

***

0.0 1.0 2.0

WTob/

ob 04 ob

Ago2mRNA(AU)

*

***

J 0 50 100 F

WTTg- 04

ob/

ob 04 ob

miR-184(AU)

0 100 200

ob/

ob Tg- 04

04 ob WT

miR-184(AU)

0 100 200

ob/

ob Tg- 04

04 ob WT

*

0 2 4 6

ob/

ob 04 ob

*

β-cell mass (mg)

WT

*

Tg- 04

*

0 2 4 6

ob/

ob 04 ob

*

β-cell mass (mg)

WT

*

Tg- 04

*

gcg+ cells/ 2mmpancreas

0 25 50

ob/

ob 04 ob WTTg-

04 n.s.

gcg+ cells/ mm2pancreas 0 25 50

0 25 50

ob/

ob 04 ob WTTg-

04 n.s.

insulin + cells/ mm2pancreas

**

0 50 100 150 200

ob/

ob 04 ob WTTg-

04

*

**

insulin + cells/ mm2pancreas

**

0 50 100 150 200

ob/

ob 04 ob WTTg-

04

*

**

body weight (g)

0 10 20 30 40 50

WTTg- 04

ob/

ob 04 ob

***

*

body weight (g)

0 10 20 30 40 50

WTTg- 04

ob/

ob 04 ob

***

*

H

n.s.

***

*** ***

Figure 4. Restoration ofmiR-184during Insulin Resistance Inhibits Compensatoryb-Cell Proliferation (A) Quantitative real-time PCR analysis ofmiR-184in islets of 8-week-old wild-type (WT),Tg-04,ob/ob, and04obmice (n = 3).

(B) Quantitative real-time PCR analysis ofmiR-375in islets of 8-week-old WT,Tg-04,ob/ob, and04obmice (n = 4).

(C) Random blood glucose of 8-week-old WT,Tg-04,ob/ob, and04obmice (n = 4–12).

(D) Plasma insulin concentrations of 8-week-old WT,Tg-04,ob/ob, and04obmice (n = 3–6).

(E) Immunostaining analysis of pancreatic sections in 8-week-old WT,Tg-04,ob/ob, and04obmice for insulin (green) and glucagon (red). Scale bars = 50mm.

(F) Pancreatic insulin content in 8-week-old WT,Tg-04,ob/ob, and04obmice (n = 4–6).

(G)bcell mass analysis of 10-week-old WT,Tg-04,ob/ob, and04obmice (n = 3).

(H) Quantification of insulin+ and glucagon+ cells per area of pancreas in WT,Tg-04,ob/ob, and04obmice (n = 4).

(I) Body weight analysis of WT,Tg-04,ob/ob, and04obmice (n = 4–12).

(J) Quantitative real-time PCR analysis ofAgo2in islets of 10-week-old WT,ob/ob, and04obmice (n = 4–5). Results presented as mean ± SEM. *p < 0.05;

**p < 0.01; ***p < 0.001.

See alsoFigure S4.

Références

Documents relatifs

contre-charme afin de les remettre en ordre croissant...

We showed that in behaving unrestrained mice performing a Go/No-Go task for odor discrimination (i) ob/ob mice were more efficient to discriminate the two odors according to

Mit ei- ner sogenannten Mediation (Vermittlung) spielte er sich zum Herrn der Schweiz auf, Hess sich feiern und sorgte einigermassen für Ruhe, indem er zwischen den

The confrontation of two bodies of academic literature, the first on migration and climate change and the second on glacial retreat impacts allows the conceptualisation

Les manifestations oto-rhino-laryngologiques ne sont pas des symptômes rares au cours du COVID-19, en particulier dans les formes légères et modérées de la maladie, de

Rhumatologie Radiologie Hématologie O.R.L Biophysique Chirurgie - Pédiatrique Chirurgie Cardio – Vasculaire Gynécologie Obstétrique Cardiologie Gastro-entérologie

Carbohydrates also cause a co- ordinated expression of metabolic enzymes in the pancreatic B-cell, which are thought to control the production of metabolic coupling

The aim of the present study was to investigate whether overexpression of the hCN1 enzyme or chronic aerobic exer- cise training, interventions potentially resulting in decreased