• Non ci sono risultati.

Human beta cell dysfunction induced by different lipoglucotoxic conditions may be transient or persistent and associates with specific transcriptomic changes which are shared in type 2 diabetes

N/A
N/A
Protected

Academic year: 2021

Condividi "Human beta cell dysfunction induced by different lipoglucotoxic conditions may be transient or persistent and associates with specific transcriptomic changes which are shared in type 2 diabetes"

Copied!
297
0
0

Testo completo

(1)

PhD Program in Clinical and Translational Science

Human beta cell dysfunction induced by

different lipoglucotoxic conditions may be

transient or persistent and associates with

specific transcriptomic changes which are

shared in type 2 diabetes

PhD Program Director

Prof. Stefano Del Prato

Tutor:

Prof. Piero Marchetti

PhD Candidate:

Mara Suleiman

(2)

KEY WORDS

Pancreatic islets

Beta-cells

Lipotoxicity

Glucotoxicity

Lipoglucotoxicity

Type 2 diabetes

Beta-cell rescue

Insulin secretion

RNA sequencing

eQTL

(3)

SUMMARY

Pancreatic beta-cell failure is determined by the interplay of genetic and acquired

factors and represent the key event that leads to the development and progression of

type 2 diabetes (T2D). The cause of this derangement is not completely clarified. Work

in vivo shows evidence that alleviation of metabolic stress, through low calorie diet,

administration of glucose-lowering drugs or bariatric surgery, can improve beta-cell

function. Studies in vitro demonstrated that prolonged exposure to saturated fatty acids

(lipotoxicity), high glucose (glucotoxicity) or combinations thereof (lipoglucotoxicity)

may contribute to beta-cell failure, possibly via endoplasmic reticulum (ER) stress,

oxidative stress, loss of identity and/or other mechanisms. In addition, the molecular

mechanisms underlying the persistence or transience of human beta-cell defects are

still to be investigated to unveil to which extent the functional and molecular

modifications (and possible recovery) in non-diabetic (ND) islets reflect those of islets

from T2D individuals.

The present thesis aimed first to assess the direct impact of different lipoglucotoxic

treatments on human beta-cell function, and then to evaluate if the deleterious effects

were persistent or reversible after washout. Finally, the associated transcriptomic

changes were analyzed and compared with T2D islet gene expression signature. Islets

obtained from 26 ND organ donors were cultured in M199 medium, containing 5.5

mmol/l glucose, for 2 days (D2); then batches of islets were cultured for 2 additional

days (D4) either in absence (ctrl) or in the presence of: 0.5 palmitate (P), 11.1 mmol/l

glucose (g), 22.2 mmol/l glucose (G), 0.5 mmol/l palmitate + 11.1 mmol/l glucose

(P+g), 0.5 mmol/l palmitate + 22.2 mmol/l glucose (P+G), 1.0 mmol/l palmitate + oleate,

(1:2 molar ratio, P+O), 1.0 mmol/l palmitate + oleate + 11.1 mmol/l glucose (P+O+g),

and 1.0 mmol/l palmitate + oleate + 22.2 mmol/l glucose (P+O+G). At D4, islets were

washed and incubated with plain M199 medium for 4 additional days (D8 control and

D8 washout, according to the incubation condition). Furthermore, 28 ND and 58 T2D

islet preparations were studied. Glucose-stimulated insulin secretion (GSIS) from all

the preparations was assessed, and islets were also prepared for RNA extraction and,

in selected cases, for histology.

No significant change occurred in GSIS with ctrl, g, P+O and P+O+g islets throughout

the study period. However, GSIS at D4 declined (p<0.05 or less) with P, G, P+g, P+G

and P+O+G exposure. Normalization of GSIS was observed at D8 washout vs D4 with

(4)

P, G and P+g, but not with P+G and P+O+G. For P, G and P+G conditions, islet

transcriptome and genome features were analyzed by RNA-sequencing and eQTL,

respectively, to unveil the molecular mechanisms underlying beta-cell damage and its

reversal. For the conditions where islets were exposed to P (functionally impaired at

D4 and rescued at D8 washout), treated islets compared to control islets at D4 had

646 differentially (FDR<0.05) expressed genes (272 up- and 374 downregulated). Of

these, 595 were protein-coding genes (248 up- and 347 downregulated by palmitate),

including genes involved in lipid metabolism, inflammation and other cell functions.

Enrichment analysis identified several P-modified functional categories including

upregulation of unfolded protein response, acyl-CoA biosynthesis and fatty acid

metabolism and others. Comparison of D8 washout vs D4 palmitate-treated islets

identified 714 genes differentially expressed (167 up- and 547 down-). Of these, 656

were protein-coding (142 up- and 514 down-) comprising genes with a role in

lipid/glucose metabolism, transcription, inflammation, beta-cell function and others.

The Enrichment Map regarding D8 washed out islets showed downregulation of fatty

acid metabolism and upregulation of carbohydrate catabolic processes. As for human

islets exposed to G (impaired GSIS at D4 with recovery after washout), the comparison

of G-exposed islets vs control islets at D4 showed 50 differentially expressed genes

(38 up- and 12 down-); among these, 42 were protein-coding (32 up- and 10

downregulated by high glucose) including genes involved in metabolic pathways,

gated channel activity and other cell functions. Enrichment analysis showed inhibition

of cell junction and metabolic processes. D8 washout vs D4 G-treated islets RNA-seq

data comparison resulted in 341 differentially expressed genes (81 up- and 260

down-regulated). Of these, 320 were protein-coding (70 up- and 250 down-), also involved

in extracellular organization, establishment of protein localization to ER and

glycerophospholipid metabolism. The relative Enrichment Map showed

downregulation of response to wounding, myeloid cell differentiation and chemotaxis.

More profound changes in islet transcriptome were observed with combined P+G

(beta-cell dysfunction at D4 and persistence after washout). D4 P+G-exposed islets

resulted in differential expression of 1,498 genes (756 up- and 742 down-). Of these,

1,386 were protein-coding (699 up- and 687 down-) and they were mainly related to

transcription, inflammation, cell turnover, ion channels/transporters, mitochondrial

function, and redox balance. The Enrichment Map showed clustering of interrelated

gene-sets upregulated for most and comprising the unfolded protein response, protein

(5)

degradation, mRNA splicing regulation and ER stress-induced apoptosis. The

RNA-seq data of human islets at P+G D8 washout vs D4 treated identified only 322 genes

differentially expressed (120 up- and 202 down-). Of them, 292 were protein-coding

(102 up- and 190 down-).

Finally, the molecular changes associated with persistent or transient beta-cell insulin

secretion defects were correlated with those of human islets from T2D donors

compared with ND donors. The Rank-Rank Hypergeometric Overlap (RRHO)

approach was used, which allows to compare differentially expressed transcriptomes

between independent experiments. Overall, the RRHO analyses showed that

persistent or transient human beta-cell dysfunction induced by metabolic stress was

accompanied by specific gene expression signatures that were shared with T2D, with

the greatest concordant overlap between conditions that induce beta-cell dysfunction

and fail to recover after washout, namely P+G and P+O+G.

These results, obtained during the PhD course, show that certain lipoglucotoxic

conditions may induce persistent or reversible beta-cell dysfunction, depending on the

type, concentration and combination of the stressors. This associates with specific

molecular changes that overlap with T2D islet traits. Identification of novel mechanisms

responsible for human beta-cell functional deterioration and rescue, which are shared

in T2D, could provide novel insights into T2D pathogenesis and should foster the

development of improved beta-cell specific therapeutic approaches.

(6)

ABBREVIATIONS

ABS

Adult Bovine Serum

ATP

Adenosine triphosphate

cis-eQTL

Cis-Expression quantitative trait loci

Ctrl

Control

DDA

Data Dependent Acquisition

dTTP

Deoxythymidine triphosphate

dUTP

2´-Deoxyuridine, 5´-Triphosphate

D2

Day 2

D4

Day 4

D8

Day 8

DPP-IV

Dipeptidyl peptidase IV

ER

Endoplasmic reticulum

eQTL

Expression quantitative trait loci

FBS

Fetal bovine serum

FCccorr

Corrected fold change

FDR

False discovery rate\

FFA

Free-fatty acids

g

11.1 mmol/l glucose

G

22.2 mmol/l glucose

GSEA

Gene Set Enrichment Analysis

GSIS

Glucose-stimulated Insulin Secretion

GWAS

Genome-wide association study

HBSS

Hanks Balanced Salt Solution

ISI

Insulin Stimulation Index

KRBS

Krebs Ringer Bicarbonate Solution

KRBH

Krebs Ringer BicarbonateHepes

LC-MS/MS

Liquid chromatography–mass spectrometry

MOI

Multiplicity of infection

mRNA

messenger Ribonucleic acid

ND

Non-diabetic

NEFA

Non-esterified fatty acids

P

0.5 mmol/l palmitate

PBS

Phosphate Buffered Saline

PBS-T0.1

Phosphate Buffered Saline, Tween 0.1%

P+g

0.5 mmol/l palmitate + 11.1 mmol/l glucose

P+G

0.5 mmol/l palmitate + 22.2 mmol/l glucose

P+O

1.0 mmol/l palmitate + oleate, (1:2 molar ratio)

P+O+g

0.5 mmol/l palmitate + 11.1 mmol/l glucose

P+O+G

0.5 mmol/l palmitate + 22.2 mmol/l glucose

qRT-PCR

Quantitative Reverse Transcription PCR

RIN

RNA Integrity Number

RIPA

Radioimmunoprecipitation assay buffer

RNA-seq

RNA sequencing

RPKM

Reads Per Kilobase of exon per Million mapped reads

RRHO

Rank-Rank Hypergeometric Overlap

SDS-PAGE Sodium Dodecyl Sulphate - PolyAcrylamide Gel Electrophoresis

siRNA

Short interfering RNA

(7)

TABLE OF

CONTENTS

KEY WORDS

i

SUMMARY

ii

ABBREVIATIONS

v

INTRODUCTION

1

PRESENT

PROJECT

7

MATERIALS AND

METHODS

8

1. Experimental model

8

1a. Human pancreatic islets

8

1b. Cell lines

9

2. Functional studies

9

2a Culture and incubation media

9

2b Culture conditions

10

2c Glucose-stimulated insulin secretion

11

3. Immunocytochemistry and electron microscopy

11

4. RNA extraction

12

5. Library preparation and sequencing

13

6. RNA-seq data analysis

14

7. Evaluation of functional enrichment

15

8. Validation studies

15

8a Proteomics and proteomic data analysis

15

8b The role of ANKRD23

17

8c The role of Sec61G

18

9. Expression quantitative trait loci (eQTL)

18

(8)

RESULTS

20

1. Impact of different lipoglucotoxic conditions on

non-diabetic (ND) islet beta-cell function

20

2. Morphometric and ultrastructural assessments

22

3. RNA-seq data of palmitate-treated human islets

22

4. RNA-seq of high glucose-treated human islets

23

5. RNA-seq of palmitate plus high glucose-treated

human islets

24

6. Comparison of palmitate, high glucose and

palmitate plus high glucose exposure

25

7. Evidence for beta-cell dedifferentiation with

lipotoxicity

25

8. Validation studies

26

9. Association with T2D genome-wide association

loci

27

10. RNA-seq of T2D vs ND human islets

28

11. Comparison of lipoglucotoxic and T2D islet gene

expression

signatures

by

Rank-Rank

Hypergeometric Overlap (RRHO)

29

CONCLUSIONS

32

TABLES AND

FIGURES

37

REFERENCES

279

(9)

INTRODUCTION

Pancreatic beta-cell failure, due to the interplay of genetic and acquired factors, is key

to the onset and progression of diabetes. Beta-cells are unique endocrine cells that

synthetize, store and secrete insulin under the control of multiple and integrated

signals, thus tightly regulating blood glucose concentrations. They have a diameter of

10 μm on average, and each of them contains approximately 20 pg insulin. Beta-cells

are the most represented endocrine cell type in pancreatic islets, comprising 50-80%

of islet cells. Studies with autoptic samples, organ donor specimens and surgical cases

have found that beta cell mass in the human pancreas may vary from 0.6 to 2.1 g, with

beta cell volume and area (relative to the pancreatic tissue) ranging 1.1-2.6% and

0.6-1.6%, respectively. In normal individuals, beta-cells release about 30–70 units of

insulin per day (essentially depending on body weight, physical activity and nutritional

habits), half of which under basal condition and the remaining in response to meals.

The most important regulator of insulin release is glucose, that acts as both a trigger

as well as an amplifier of insulin secretion. Several other physiological molecules

regulate insulin secretion, including non-carbohydrate nutrients, hormones and

neurotransmitters.

In type 2 diabetes (T2D), the most common form of diabetes (representing

approximately 80-90% of all cases), beta-cell incompetence is due to both reduced

mass and functional impairment. Studies in-vitro and in-vivo have recently proposed

that dysfunction, rather than death, is the prevalent defect of the beta-cells in T2D.

Since the UK Prospective Diabetes Study [1], it has been assumed that the decline of

beta-cell functional mass begins before the onset of T2D and proceeds relentlessly

thereafter, leading to worsening of glycemic control and requiring progressive

intensification of diabetes therapy. The causes of this deterioration are not completely

understood, but prolonged exposure to saturated fatty acids (lipotoxicity), high glucose

(glucotoxicity) or combinations thereof (lipoglucotoxicity) have been proposed to

contribute to beta-cell failure both in vivo and ex vivo, probably via endoplasmic

reticulum (ER) stress, oxidative stress, loss of identity and other mechanisms [2–13].

Of interest, growing evidence shows that alleviation of metabolic stress can improve

beta-cell function and even induce remission of T2D. In the 1990’, a study was

performed aimed to investigate short- and long-term effects of lipid infusion on insulin

secretion. Twelve healthy individuals underwent a 24-h Intralipid (10% triglyceride

(10)

emulsion) infusion. After an overnight fast (baseline), at 6 and at 24 h of i.v. Intralipid

administration and 24 h after Intralipid discontinuation (recovery test), all subjects

underwent an intravenous glucose tolerance test. Intralipid infusion caused a threefold

increase of circulating plasma non-esterified fatty acid (NEFA) concentrations, with no

difference between the 6- and the 24-h time points. Compared to baseline acute insulin

response (AIR), short-term (6-h) Intralipid infusion was associated with a significant

increase of insulin secretion, whereas long-term (24-h) Intralipid administration

induced a significant reduction of AIR. The recovery test showed that after 24h from

Intralipid infusion discontinuation fasting plasma NEFA concentrations and AIR values

had returned to baseline values, demonstrating reversibility of beta-cell functional

alterations induced by the “lipotoxic” condition in vivo [14].

A few years later, insulin secretion was evaluated during a 4-day lipid infusion in normal

glucose tolerant individuals with or without a family history positive for T2D [15].

Thirteen and 8 subjects in the first and second group were studied, and received in

random order a lipid (Liposyn III, 20% triglyceride emulsion) or saline infusion. On days

1 and 2 insulin and C-peptide levels were measured after standardized mixed meals,

and a hyperglycemic clamp was performed on day 3. Similar concentrations of NEFA

were observed in the two groups during the study. In control subjects, the lipid infusion

determined a significant increase of insulin secretion after mixed meals and during the

hyperglycemic clamps. On the contrary, beta cell function decreased markedly in the

group with T2D family history, which affected both first and second phase insulin

release. These alterations became even more evident after correction for insulin

sensitivity. The results indicated therefore that subjects who are genetically at risk to

develop T2D, are particularly susceptible to the beta-cell functional damage induced

by increased plasma fatty acids (lipotoxicity), resulting in reduced insulin secretion in

response to mixed meals and intravenous glucose challenge. In a follow-up study, the

same group assessed the effects of pharmacological reduction of NEFA in 9

non-diabetic volunteers with a strong predisposition to T2D [16]. They used Acipimox, an

antilipolytic nicotinic acid derivative. Subjects were admitted twice and received in

random order either the drug or placebo for two days, in a double-blind design. In

addition to plasma glucose, insulin and C-peptide measurements during the days, a

hyperglycemic clamp was performed on day 3. Acipimox reduced 48-h plasma FFA by

about one third, which was associated with improved beta-cell function during the day.

First and, more evidently, second-phase insulin secretion during the hyperglycemic

(11)

clamp also improved, which was particularly remarkable after adjustment for the

prevailing insulin resistance. This study and its results provide further evidence for the

relevant role of lipotoxicity in the impairment of beta-cell function, at least in individuals

predisposed to T2D. More importantly, they demonstrate that beta-cell functional

damage can be rescued, provided the metabolic insult is attenuated.

If and how the possible recovery of beta-cell function observed in non-diabetic

individuals may apply to patients with overt T2D and impact on diabetes clinical

trajectory in still matter of debate. Remission of diabetes of variable duration can be

achieved in proportions of T2D subjects by carbohydrate restriction, low-calorie diets,

pharmacological treatments and bariatric surgery [17–22]. A few studies have

assessed if changes at the beta-cell level play a role in T2D remission following

low-calorie diets and bariatric surgery. In an early study [23] eleven subjects with T2D were

examined before and after 1, 4 and 8 weeks of a low-calorie (600 kcal)/day) diet. The

results showed that after 1 week of restricted energy intake fasting plasma glucose

normalized in the diabetic group. The first-phase insulin secretion increased during the

study period and approached control values. Maximal insulin response became

supranormal at 8 weeks. This was accompanied by significant decrease of pancreatic

triacylglycerol content. In a more recent article [24] detailed metabolic studies were

performed in subgroups of the Diabetes Remission Clinical Trial (Direct), in which

remission of T2D and persistence of non-diabetic blood glucose control were achieved

in 46% of patients treated with a low-calorie diet [17]. This specific study included 64

and 26 people respectively from the intervention and control groups). The authors

found that pancreatic fat content decreased whether or not glycemic values

normalized. However, recovery of first-phase insulin release identified those

individuals with remission of diabetes, which was durable at one year. These results

demonstrated that beta cell function can be rescued in a sizeable proportion of T2D

subjects, to achieve and sustain normoglycemia.

In the case of bariatric surgery, there is evidence that Roux-en-Y gastric bypass

(RYGB), vertical sleeve gastrectomy (VSG) and biliopancreatic diversion (BPD)

improve glucose control and can promote remission of T2D independently from weight

reduction [25,26]. The beneficial effects on glycemic indices may persist over the years

in several patients and become more evident as weight loss progresses [27–30].

Although bariatric surgery has distinct effects on several organs and functions,

post-surgical subjects with previous T2D show rapid and definite improvements of insulin

(12)

secretion, that persist over the time [31–33]. Restoration of first-phase insulin release

to intra-venous glucose administration (one to four weeks after RYGB or BPD) has

been consistently observed, occurs before any significant weight loss and reasonably

contributes to diabetes improvement or remission after the bariatric surgery procedure

[34–36]. Although the mechanisms determining better beta-cell function after bariatric

surgery are still largely unclear, increased GLP-1 release and potentiated incretin

effect are believed to play a major role [37,38].

In the past few years some information has been generated on the reversal of

beta-cell damage directly from isolated human islets, under different experimental

conditions. The use of human islets, prepared from the pancreas of organ donors,

allow assessment of beta-cell features independently from biological factors that

in-vivo could confound the scenario, and, importantly, can shed light on islet cell

morphological and molecular traits.

In an early study, [39] islets prepared from seven ND human pancreata were cultured

for 48h in normal or high glucose (16.7 mmol/l) containing medium. Islets were then

perifused with different glucose concentrations (3.3 and 16.7 mmol/l) or 10 mmol/l

L-arginine. It was seen that islets cultured in the high glucose medium lost acute

glucose-induced insulin release. However, insulin release in response to arginine was

preserved, supporting the concept of a selective loss of beta-cell sensitivity to glucose

stimulation induced by glucotoxicity. Notably, after additional 48h culture in normal (at

5.5 mmol/l glucose concentration) medium, the islets partially recovered from their

insulin secretion dysfunction. A few years later, these findings were confirmed and

implemented in a study in which ND islets were exposed to 33 mmol/l glucose for four

and nine days [40]. After these glucotoxic incubations, the authors observed a severe

decrease of insulin content (that was correlated with the reduction of insulin mRNA

and the transcriptional activity of PDX1) and the loss of glucose-stimulated insulin

release. Interestingly, most of these beta-cell alterations were partially reversible when

islets previously cultured for 6 days in high glucose were transferred to normal glucose

(5.5 mmol/l) concentrations for 3 additional days.

A few studies have assessed whether the defects of islets prepared from T2D donors

can be counteracted. The first data on this issue were generated with six T2D islet

preparations [41]. The authors found that, compared to ND islets, T2D islets showed

reduced insulin content, decreased amount of mature insulin granules, impaired

glucose-induced insulin secretion, reduced insulin mRNA expression, increased

(13)

apoptosis, together with higher expression of nitrotyrosine (a marker of oxidative

stress) and genes involved in the redox balance. Remarkably, the study showed that

24h incubation of T2D islets with a therapeutical concentration of metformin was

associated with increased insulin content, increased number and density of mature

insulin granules, improved glucose-induced insulin release, increased insulin mRNA

expression and reduced apoptosis. These effects were associated with decreased

oxidative stress, as suggested by lower levels of nitrotyrosine and reduced expression

of NADPH oxidase, catalase and GSH peroxidase after exposure to metformin.

On a similar line, some evidence has been provided to show that molecules of the

incretin system may have direct beneficial effects on T2D islets. In a study with islets

from seven T2D and eleven ND donors, the cells were exposed for 48h to 10 nmol/l

exendin-4, a DPP-4 resistant GLP-1 mimetic [42]. The authors observed that

incubation with exendin-4 caused a significantly better glucose-stimulated insulin

release from both T2D and ND islets, in comparison with untreated cells. In diabetic

islets this was associated with increased expression of a few key beta-cell genes,

including glucokinase, PDX-1, E2F1 and Cyclin D1.

A couple of years later the same group assessed the direct effect of GLP-1 and GIP

(alone or in combination) on isolated human T2D (four donors) and ND (eleven donors)

islets. Islets were exposed to incretins for 45 min (acute exposure: 0.1, 1.0, 10 or 100

nmol/l) or 2 days (prolonged exposure: 10 nmol/l), and functional as well as molecular

features were then assessed. After acute exposure (at concentrations from 1.0 to 100

nmol/l), both GLP-1 and, more markedly so, GIP, significantly improved

glucose-stimulated insulin release in ND islets, with no apparent synergic action. Similar effects

were observed with T2D islets acutely treated with 10 nmol/l GLP-1 or 100 nmol/l GIP.

Following prolonged exposure, improved insulin secretion was observed with T2D

islets cultured with GIP and GLP-1 in combination with GIP. Transcription of insulin,

PDX-1 and Bcl-2 tended to be higher after incretin culture in both T2D and ND islets,

with the combination of GLP-1 and GIP showing more significant effects.

Finally, more recent work [43] investigated the effects of compounds involved in the

modulation of autophagy (a process that leads to the degradation and recycling of

intracellular components) on beta-cell function, survival and ultrastructure. Islets

isolated from seventeen ND and nine T2D organ donors were cultured for 1–5 days

with 10 ng/ml rapamycin (an autophagy inducer), 5 mM 3-methyladenine (3-MA) or 1.0

nM concanamycin-A (autophagy blockers), either in the presence or not of metabolic

(14)

(0.5 mM palmitate) or chemical (0.1 ng/ml brefeldin A) endoplasmic reticulum (ER)

stressors. In ND islets glucose-stimulated insulin secretion was reduced by palmitate

and brefeldin; rapamycin prevented palmitate-, but not brefeldin-mediated cytotoxic

damage. Palmitate (and in similar way, brefeldin A) exposure increased beta-cell

apoptosis in ND islets, which was significantly prevented by rapamycin and worsened

by 3-MA. Both palmitate and brefeldin induced the expression of ER stress markers

(PERK, CHOP BiP) which was partially, but significantly prevented by rapamycin. In

T2D islets, the exposure to rapamycin improved insulin secretion, reduced the

proportion of beta-cells with signs of apoptosis and preserved insulin granules,

mitochondria and ER ultrastructure compared to untreated T2D islets; this was

associated with a significant reduction of PERK, CHOP and BiP gene expression.

These findings are in line with the concept that supporting autophagy can improve

human beta-cell function and survival under ER stress conditions and in the case of

T2D.

All these in-vivo and ex-vivo data support the concept that beta-cell dysfunction

induced by metabolic stress and in the case of T2D may be rescued, at least in part.

However, extensive assessments of these issues are lacking, and very little is known

on the associated molecular features.

In this PhD thesis a comprehensive study was performed with a large number of human

islet preparations to assess the direct impact of different lipoglucotoxic treatments on

beta-cell function and evaluated if the deleterious effects were persistent or reversible

after washout. The ex-vivo lipoglucotoxic treatments were selected to reflect

biologically relevant concentrations of the most common saturated and

mono-unsaturated fatty acid and glucose. For key conditions islet transcriptome and genome

were analyzed by RNA-sequencing and eQTL to examine the possibly involved

molecular mechanisms. Finally, the molecular changes associated with persistent or

transient beta-cell dysfunction were correlated with those of islets from T2D donors

compared with ND subjects. Mechanisms associated with human beta-cell functional

deterioration and rescue were identified, and their overlap with T2D islet traits unveiled,

to provide insights into T2D pathogenesis and foster the development of improved

β-cell targeted therapeutic strategies.

(15)

PRESENT PROJECT

With this project I present a comprehensive study (Figure 1) with a large series (n=26)

of ND human islet preparations (Table 1) in which the direct impact of several

lipoglucotoxic treatments on beta-cell function was assessed, aiming to evaluate if the

deleterious effects were persistent or reversible after the removal of the stressors

(washout). The ex vivo lipoglucotoxic treatments were selected to reflect biologically

relevant concentrations of the most common saturated and mono-unsaturated fatty

acid and glucose [44]. For specific treatments following which persistence of the

damage or recovery was observed, islet transcriptome and genome were analyzed by

RNA-sequencing and eQTL to shed light on the molecular mechanisms possibly

involved. In addition, the molecular changes associated with persistent or transient

beta-cell dysfunction were correlated with those found by comparing the transcriptomic

profile of islets from 28 T2D donors with 58 ND subjects (Tables 2a, 2b), to provide

insights into T2D pathogenesis and possibly, towards the development of therapeutic

strategies beta-cell specific.

(16)

MATERIALS AND METHODS

1. Experimental models

1a. Human pancreatic islets

Islets were prepared from the pancreas of 26 ND organ donors (16 females, 10 males,

age: 72±3 years, BMI: 24.6±0.7 Kg/m

2

) [45,46] at the Pancreatic Islets Laboratory, with

permission by the Ethics Committee of the University of Pisa, upon written consent of

donors’ next-of-kin. The main clinical characteristics of each donor are provided in

Table 1 together with features of the processed pancreases and isolated islets. In a

separate set of experiments, islets were obtained from the pancreas of 28 T2D donors

(9 females and 19 males, age: 73.6±1.5 years, BMI: 26.3±0.7 Kg/m

2

) and 58 ND (30

females and 28 males, age: 64.4±2.1 years, BMI: 25.3±0.5 Kg/m

2

) (detailed

information is provided in Table 2a, 2b). Islets were isolated by enzymatic digestion

and density gradient purification; briefly, the gland was cleaned by careful dissection

of the surrounding fat tissue, duodenum portion, lymph nodes, vessels and spleen (if

present). After removing the head of the organ to access the Wirsung duct, body and

tail portion was used for islet preparation. The pancreatic duct was cannulated with a

18-gauge angio catheter and 200 ml of Hank’s Balanced Salt Solution (HBSS,

Sigma-Aldrich MO - USA), completed with 10% Adult bovine serum (ABS, Sigma-Sigma-Aldrich) and

3.6 mg/ml collagenase P (Roche Diagnostics, Switzerland) was injected into the

pancreatic duct until it completely distended the gland. Then, the pancreas was placed

into a sterile glass beaker in a water bath (37°C for 10 minutes). Successively, the

gland was gently hand-shaken at room temperature until free islets were observed in

a sample. At this point, the digestate was sequentially passed through 300-μm and

90-μm mesh stainless steel filters. The resulting suspension and the tissue trapped on the

300-μm mesh filter were placed back into the beaker with HBSS for further digestion.

The tissue remaining on the 90-μm filter was washed with HBSS supplement with 10%

of ABS. The same procedures of filtration, washing, and settling in the HBSS solution

was repeated at 5- to 8-minute intervals up to 40 minutes. For the purification, the

digested suspension was aliquoted in to 50-ml Falcon conical tubes and centrifuged at

1000 rpm for 2 minutes. The supernatant was discarded and the pellet suspended in

15 ml of a solution containing Lympholyte (Euro-Clone, Milano – Italy): HBSS (with

(17)

10% ABS) with different ratio (for example 80:20 or 70:30). Then 10 ml of HBSS was

layered over the Lympholyte-HBSS solution. The tubes were centrifuged at 1800 rpm

for 5 minutes at 4°C. Islets recovered from the interface between the two layers were

again centrifuged at 1800 rpm for 2 minutes, following which the supernatant was

discarded and the islet pellets were aliquoted into 75-cm

2

Corning® cell culture flasks

(Sigma-Aldrich) containing complemented M199 medium (Euro-Clone); the medium

was refreshed after 24h culture. The islet yield was evaluated by staining a sample

with dithizone (DTH, Sigma-Aldrich). The purity of the islets was calculated as the ratio

between DTH-stained and total (stained and unstained) cell clusters [47].

1b. Cell lines

For validation studies, three different beta cell-lines were used. The EndoC-βH1 cell

line, a beta-cell line derived from human fetal pancreas, was cultured in DMEM

containing 5.6 mmol/l glucose, 2% BSA fraction V, 50 µmol/l 2-mercaptoethanol

(Sigma-Aldrich, Poole, UK), 10 mmol/l nicotinamide (Calbiochem, Darmstadt,

Germany), 5.5 µg/ml transferrin, 6.7 ng/ml selenite (Sigma-Aldrich), 100 units/ml

penicillin and 100 µg/ml streptomicin (Lonza, Leusden, The Netherlands). The same

medium, but with 2% FBS, was used after transfection with siRNAs. INS-1E beta-cells,

were cultured in RPMI 1640, containing 11 mmol/l glucose, 10% fetal bovine serum,

10 mmol/l HEPES, 50 μM 2-mercaptoethanol, and 1 mmol/l sodium pyruvate at 37°C

with 5% CO2 in a humidified atmosphere. MIN6B1 cells were cultured in 25 mmol/l

glucose DMEM supplemented by 15% fetal bovine serum, 2 mmol/l L-glutamine, 20

mmol/l HEPES, 50 μmol/l β-mercaptoethanol, plus penicillin (100 units/mL) and

streptomycin (0.1 mg/mL) at 37˚C in an atmosphere of humidified air (95%) and CO2

(5%).

2. Functional studies

2a. Culture and incubation media

The culture medium (M199 medium) was prepared from M199 medium (Euro-Clone)

containing 5.5 mmol/l glucose, supplemented with 0.1 mg/ml L-glutamine (Sigma

Aldrich), 10% ABS (Sigma-Aldrich), 100 UI/l penicillin (Sigma-Aldrich), 100 mg/l

streptomycin (Sigma-Aldrich), 750 μg/l amphotericin B (Sigma-Aldrich) and 50 mg/l

(18)

gentamycin (Sigma-Aldrich). pH was adjusted to 7.35-7.40 and the final solution was

filtered with a 0.2 Vacuum filter, PES (Sarstedt, Germany). The palmitate and the

oleate incubation media were obtained by adding the free-fatty acids (FFA) solutions

to M199 medium. FFA solutions were obtained by dissolving palmitate (sodium salt,

Sigma-Aldrich) and oleate (sodium salt, Sigma-Aldrich) in 90% volume/volume

ethanol, heated to 60°C and 1:100 diluted to a final concentration of 0.5 mmol/l.

Palmitate was used at a molar ratio of palmitate:bovine serum albumin of 3.3 [48]. The

11.1 mmol/l and 22.2 mmol/l glucose incubation media were prepared by dissolving

5.5 mmol (990 mg) and 16.7 mmol (3,006 mg) glucose to 1L M199 culture medium.

The Krebs Ringer Bicarbonate Solution (KRBS), used for insulin secretion studies was

prepared by combining equal volumes of 4 separate solutions:

Solution 1: KRBS containing 0.5 mol/l NaCl

Solution 2: KRBS containing 20 mmol/l KCl, 96 mM NaHCO

3

, 4 mM MgCl

2

•6H2O.

Solution 3: KRBS containing 10 mmol/l CaCl

2

Solution 4: KRBS containing 2% Bovine Serum Albumin Fraction V (BSA, Roche

Diagnostics, Germany)

2b. Culture conditions

After the isolation, around 2000 ND islets were cultured in M199 medium [46,49] for 2

days (D2), to allow recovery from the isolation process. Then, based on previously

reported procedures [10,45,49], batches of islets were cultured for 2 additional days

(D4) under one of the following metabolically stressful conditions: 0.5 palmitate (P),

11.1 mmol/l glucose (g), 22.2 mmol/l glucose (G), 0.5 mmol/l palmitate + 11.1 mmol/l

glucose (P+g), 0.5 mmol/l palmitate + 22.2 mmol/l glucose (P+G), 1.0 mmol/l palmitate

+ oleate, (1:2 molar ratio, P+O), 1.0 mmol/l palmitate + oleate + 11.1 mmol/l glucose

(P+O+g), and 1.0 mmol/l palmitate + oleate + 22.2 mmol/l glucose (P+O+G). Islet

batches from the same donor were also cultured in normal M199 medium in parallel.

At D4, both control islets and treated islets were washed and incubated with M199

medium for 4 additional days (indicated respectively as D8 control and D8 washout)

(schematically represented in Figure 1A). The number of separate islet preparations

per condition ranged from 3 to 6 and replicates for each series ranged from 26 to 54,

for a total number of 277.

(19)

ND and T2D islets from separate experiments were cultured in normal M199 medium

for 2-3 days, before the experimental procedures, to allow recovery from the islet

preparation process (Figure 1B).

2c. Glucose-stimulated insulin secretion

Insulin secretion in response to acute glucose stimulation was assessed with

handpicked islets by batch incubation, and islet insulin content was measured after

acid-alcohol extraction [46,50]. Fifteen islets of similar size (approximate diameter 150

mm) were hand-picked and pre-incubated with KRBS containing 3.3 mmol/l glucose

(2 ml) at 37°C for 45 min, followed by a short wash and an incubation with 3.3 mmol/l

glucose (2 ml) at 37°C for 45 min (basal). The islets were then challenged with 16.7

mmol/l glucose (2 ml) at 37°C for additional 45 min (stimulated). After centrifuge step,

the respective supernatants were collected for insulin quantification; the bottom islet

pellets were incubated with an acid-alcohol solution (2 ml) at 4°C overnight for

subsequent insulin content measurement. Basal and stimulated insulin release, and

insulin content were quantified by a radioimmunometric assay (DIAsource

ImmunoAssays S.A., Nivelles, Belgium).

Insulin release was expressed as actual and normalized (to insulin content) values

respectively; insulin stimulation index was calculated as ratio of insulin release at 16.7

mmol/l glucose over release at 3.3 mmol/l glucose.

3. Immunocytochemistry and electron microscopy

Isolated islets, from selected samples, were studied by light microscopy

immunocytochemistry

[45]

and/or

electron

microscopy

[52-53].

For

immunocytochemistry, islet pellets were first 4% paraforlmaldehyde-fixed at room

temperature for 1 h, then washed with 1X-phosphate buffered saline (PBS,

Sigma-Aldrich) and resuspended in HistoGel

TM

(Thermo Scientific, Waltham, MA, USA), a

liquid embedding medium used to encapsulate and retain small and friable specimens.

After the complete solidification of the gel containing the islets, the block was handled

according to tissue processing and paraffin-embedding [51]. Then, sequential 2 mm

sections were cutted from the gel block; hematoxylin-eosin staining was performed to

assess the section quality; sequential sections were stained by applying the following

(20)

primary antibodies: guinea pig anti-insulin antibody, (1:100, Invitrogen, Carlsbad, CA,

USA); polyclonal rabbit anti-human glucagon antibody, (1:3,000, Dako, Carpinteria,

CA, USA); mouse monoclonal anti-chromogranin A antibody, (1 μg/ml, Ventana, Oro

Valley, AZ, USA). Biotinylated secondary antibody, which reacts with mouse, rabbit,

guinea pig and rat primary antibodies, was purchased as Histostain-Plus kits

(Invitrogen). Sections were analyzed using Leica DM5500 B microscope (Leica,

Wetzlar, Germany) equipped with the DFC310 FX camera (Leica). Images were

acquired using Leica HCX PL FLUOTAR objective lenses at 40X magnification and

processed by the Leica MetaMorph® software, v1.8.0.

For electron microscopy, 30 hand-picked islet pellets were fixed with 2.5% (vol./vol.)

glutaraldehyde in 0.1 mmol/l cacodylate buffer, pH 7.4 for 1h at 4°C, and then postfixed

in 1% (vol./vol.) cacodylate-buffered osmium tetroxide for 2 h at room temperature.

Samples were dehydrated in a graded series of ethanol, transferred to propylene oxide

and embedded in Epon-Araldite. Ultrathin sections (60–80 nm thick) were cut with a

diamond knife, placed on formvar/carbon-coated copper grids (200 mesh), and stained

with uranyl acetate and lead citrate [52,53]. Apoptotic beta-cells were identified based

on the presence of marked chromatin condensation in the nucleus and/or blebs, as

described by Masini et al. 2009, Galluzzi et al. 2007 [52,54].

4. RNA extraction

For the lipoglucotoxic experiments, approximately 120 islets per condition were used.

They were handpicked, washed with sterile PBS (Sigma-Aldrich), centrifuged at 3,000

× g for 5 min to remove the washing buffer, snap frozen and then stored at -80 °C until

RNA extraction [46,55]. Total RNA was prepared using the RNeasy MINI Kit +

QIAshredder (Qiagen, Hilden, Germany); samples were lysed and homogenized in the

presence of a highly denaturing guanidine-thiocyanate-containing buffer which

immediately inactivates RNases. Ethanol was added to provide total RNA appropriate

binding conditions to the membrane of the speed column. Genomic DNA

contamination was removed by treating samples with DNase I (RNase-Free DNase

Set; Qiagen GmbH, Hilden, Germany); each sample was incubated with 80 ml of

solution containing 10 ml DNase I (27 units) + 70 ml buffer RDD at room temperature

for 15 min. Consecutive washing steps were finally performed, RNA was eluted in 30

ml water and concentration and integrity measurements were assessed.

(21)

RNA extraction from ND and T2D islets was performed using 100–120 islets, rinsed in

sterile PBS (Sigma-Aldrich) and centrifuged at 3,000 × g for 5 min to. The washing

PBS was disposed and 100 μl of extraction buffer (PicoPure RNA Isolation Kit; Life

Technologies, Foster City, CA) was added. Samples were incubated at 42°C for 30

min, centrifuged at 3,000 × g for 2 min, and the supernatant was collected and stored

at −80°C until RNA isolation. RNA isolation was performed according to the

manufacturer’s protocol. Briefly, 100 μl of 70% ethanol were added to the cell extract,

the mixture was added to the purification columns, washed, and subjected to DNase

treatment by incubation with 40 μl of DNase I solution (RNase-Free DNase Set, Qiagen

GmbH) for 15 min. Two additional washes were performed and RNA was eluted in 30

μl of elution buffer. RNA concentration (absorbance 260) and purity were evaluated

using the NanoDrop™ 2000c Spectrophotometer (Thermo Fisher Scientific) which

allowed the use of minimum volume-size, and provided information on purity utilizing

spectral output to detect possible protein (absorbance 280) or phenols (absorbance

230) contamination. RNA quality was assessed using Agilent Bioanalyzer 2100

(Agilent Technologies, Wokingham, UK) and Agilent RNA Nano Chips (Agilent

Technologies). The RNA integrity number (RIN), provided by the instrument, is based

on an algorithm which extracts the relationship between different characteristics of the

measurement. RIN values from the samples of the lipoglucotoxic series were: mean

8.9; SEM 0.1; median 8.8, range 7.9–9.7; the RIN values from ND and T2D samples

used for the T2D vs ND islet comparison had a RIN value of 7.5 and above. All the

samples had RIN values ensuring suitability for sequencing.

5. Library preparation and sequencing

Libraries were prepared using the TruSeq Stranded mRNA Library Prep Kit (Illumina,

San Diego, CA, USA). Briefly, mRNA was purified from 0.40 µg total RNA using

poly-T oligo attached magnetic beads. Following purification, mRNA was fragmented using

divalent cations and exposure at 94°C for 1 min, followed by 4°C hold temperature, in

order to obtain insert sizes ranging from 130 and 310 nucleotides in length. Cleaved

RNA fragments were copied into first strand cDNA using reverse transcriptase and

random primers. Second strand cDNA synthesis was performed by replacing dTTP

with dUTP in the second strand, using DNA Polymerase I and RNase H to generate

blunt-ended ds cDNA. The incorporation of dUTP quenches the second strand during

(22)

amplification. Adenylation of the 3’ ends of the blunt fragments and adaptor ligation

were performed to prepare the ds cDNA for hybridization onto a flow cell. Selective

enrichment of DNA fragments with adapter molecules on both ends and DNA

amplification of the first strand were obtained by PCR. The polymerase used does not

amplify the second strand incorporating dUTP. The generated libraries were quantified

using qPCR and quality was assessed using Agilent Bioanalyzer 2100 (Agilent

Technologies) and Agilent DNA 1000 chips (Agilent Technologies). All the libraries had

concentration and size (300 bp) suitable for sequencing which was performed on the

Illumina HiSeq 2500 instrument (Illumina, Inc. San Diego, California, U.S) for the

lipoglucotoxic conditions, and on an Illumina Genome Analyzer II (Illumina) for the T2D

and ND samples (Figure 2A). Paired-end sequencing (2x100bp) at 170 million reads

was performed.

6. RNA-seq data analysis

Sequenced reads from lipo/glucotoxicity experiments were mapped to the human

genome (assembly version GRCh37/hg19) using TopHat 2 (v2.0.13) [56] with default

parameters (Figure 2B, C). Reads were assigned to their corresponding exons and

genes based on GENCODE annotation v18 [57] using Flux Capacitor [58] with default

parameters. Resulting raw gene counts were normalized to gene length (sum of exons)

and sequencing depth, i.e., reads per kilobase per million (RPKM) mapped reads

(Figure 2D). The differentially expressed genes were identified by the R/Bioconductor

package EdgeR [59] using the raw counts with a false discovery rate (FDR) <0.05 as

cut-off for significance. The p-value for EdgeR differential analysis were corrected

according to the Benjamini and Hochberg 1995 method, as implemented by R p.adjust

function [60] (Figure 2E). For the comparison of differential expression of

lipo/glucotoxicity versus T2D, sequence reads were quantified with Salmon 13 [61]

using the transcriptome GRCh38 version 95 in quasi-mapping mode with sequence

and GC bias corrections and mapping validation. Differential analysis was done using

R DESeq2 package version 1.24.0 [62]. The enrichment map and gene list and plots

were generated by a modified two-tailed Rank-Rank Hypergeometric Overlap (RRHO)

method [63,64]. The logarithm of fold change was used to rank differentially expressed

genes. For the within-group comparisons over time, time-induced effects were first

evaluated by performing differential transcription analysis for the control conditions

(23)

between D4 vs D8, which reflects time-induced effects in the absence of a metabolic

insult. Next, the number of differentially expressed genes common to treatment and

control, and the magnitude of change over time were examined. Differentially

expressed genes with larger fold change of expression by treatment than control/time

were considered as “true” differentially expressed genes. The time effect was also

taken in to account in the RRHO analyses, through computing a corrected fold change

(FCcorr). From the fold change of D4 P±G (FCa) vs D4 control and D8 P±G vs D8

control (FCb), FCcorr is given by FCb/FCa. The RRHO software was modified to better

take in to account the multiplicity of minimal p-values, null p-values and the asymmetry

between the number of genes up- or down-regulated in the two datasets. The p-value

for RRHO was controlled for multiple testing with the Benjamini and Yekutieli method

(2001) as implemented by R p.adjust function [65]. The functional enrichment analysis

for the RRHO gene lists was generated with the gprofiler2 R package [66].

7. Evaluation of functional enrichment

Functional enrichment was generated using Gene Set Enrichment Analysis (GSEA)

softwar0e v3.0 [67]. Standard parameters were applied, except for minimum and

maximum size of functional category values that were adjusted to 5 and 500,

respectively. Enrichment maps of significantly modified biological processes (Gene

Ontology) were generated using the plugin “Enrichment Map” v3.1 [68] and visualized

within Cytoscape v3.6, using as geneset similarity cutoff a Jacard Index of 0.25 and a

FDR <0.05. To identify genes most likely to be relevant to the clusters of genesets

produced by Enrichment Mapping, the genes most frequently appearing across their

leading-edge analysis were identified [67] (Figure 2F). The leading-edge subset

represents the core of genes accounting for the gene set’s enrichment signal in GSEA.

Heatmaps show GSEA rank metric scores from these genes; higher absolute values

identify critical genes (Figure 2F).

8. Validation studies

8a. Proteomics and proteomic data analysis

Proteomics experiments [69] were accomplished with human islets to verify the protein

expression of some of the differentially expressed genes; functional as well as

(24)

molecular experiments were conducted to validate the role of genes associated with

changes in insulin secretion in the human islet incubation conditions described above.

For shotgun proteomics analysis, experiments were performed similar to what

previously described [69–72]. Briefly, islets were collected and washed twice with PBS

(37 °C). Cells were suspended in the buffer solution (7 M urea, 2 M thiourea, 4%

CHAPS, 60 mM dithiothreitol (DTT), 0.002% bromophenol blue) added with 50 mM

NaF, 2 mM Na3VO4, 1 μL/106 cells protease inhibitors, 1 µM trichostatin A, 10 mM

nicotinamide. After stirring and sonication (4 seconds, 5 times) cells were allowed to

rehydrate for 1 h at room temperature with occasional stirring. Thereafter, the solution

was centrifuged at 17,000 g for 5 min at room temperature. Protein concentration of

the resulting supernatant was determined using the Bio-Rad RC/DC-protein assay

(Bio-Rad). BSA was used as a standard. [69]. Aliquots of 40 µg of human islets protein

extracts were loaded onto 12% acrylamide resolving gel and subjected to

1D-electrophoresis. After protein visualization by Comassie blue, gel bands (16 bands for

lane) were excised, destained and subjected to in-gel reduction, alkylation and

overnight trypsin digestion at 37°C. The resulting peptides were analyzed in technical

triplicates by LC-MS/MS using a Proxeon EASY-nLCII (Thermo Fisher Scientific,

Milan, Italy) chromatographic system coupled to a Maxis HD UHR-TOF (Bruker

Daltonics GmbH, Bremen, Germany) mass spectrometer equipped with a nanoESI

spray source. Peptides were loaded on the EASY-Column C18 trapping column (2 cm

L., 100 µm I.D, 5 µm ps, Thermo Fisher Scientific), and subsequently separated on an

Acclaim PepMap100 C18 (75 µm I.D., 25 cm L, 5 µm ps, Thermo Fisher Scientific)

nano scale chromatographic column. The flow rate was set to 300 nL/min and the

gradient was from 3 to 35% of B in 80 min followed by 35 to 45% in 10 min and from

45 to 90% in 11 min. Mobile phase A was 0.1% formic acid in H2O and mobile phase

B was 0.1% formic acid in acetonitrile. The mass spectrometer was operated in positive

ion polarity and Auto MS/MS mode (Data Dependent Acquisition - DDA), using N2 as

collision gas for CID fragmentation. Precursors in the range 350 to 2,200 m/z

(excluding 1,220.0–1,224.5 m/z) with a preferred charge state +2 to +5 (excluding

singly charged ions) and absolute intensity above 4,706 counts were selected for

fragmentation in a maximum cycle time of 3 seconds. After acquiring one MS/MS

spectrum, the precursors were actively excluded from selection for 30 seconds.

Isolation width and collision energy for MS/MS fragmentation were set according to the

mass and charge state of the precursor ions (from 3 to 9 Da and from 21 eV to 55 eV).

(25)

In-source reference lock mass (1,221.9906 m/z) was acquired online throughout the

runs.

For bioinformatics processing, raw data were processed using PEAKS Studio v7.5

software (Bioinformatic Solutions Inc, Waterloo, Canada) using the ‘correct precursor

only’ option. The resulting mass lists were searched against nextprot database

(including isoforms as of June 2017; 42,151 entries). Carbamidomethylation of

cysteines was selected as fixed modification, oxidation of methionines and

deamidation of asparagine and glutamine were set as variable modifications.

Non-specific cleavage was allowed to one end of the peptides, with a maximum of 2 missed

cleavages. The highest error mass tolerances for precursors and fragments were set

at 10 ppm and 0.05 Da, respectively.

8b. The role of ANKRD23

To assess the effect of Ankrd23 overexpression on AMPK activation, the MIN6B1 cells

were plated on a 12-well plate, in duplicate for each condition and infected by an

adenovirus expressing either GFP (control) or mouse Ankrd23 (Vector Biolabs,

Malvern, PA, USA) at a multiplicity of infection (MOI) of 10 virus particle/cell. Two days

post-infection, the cells were preincubated for 1h in Krebs Ringer Bicarbonate Hepes

(KRBH) buffer (40 mM NaCl, 3.6 mM KCl, 0.5 mM NaH2PO4, 0.2 mM MgSO4, 1.5 mM

CaCl2, 10 mM HEPES, 25 mM NaHCO3), saturated with 95% O2/5% CO2 and

adjusted to pH 7.4 with the addition of 3 mM glucose before incubation for 30 min in

low (3 mM) or high (30 mM) glucose. The cells were then lysed in 100 μL

Radioimmunoprecipitation assay buffer (RIPA) containing protease (cOmplete™

Protease Inhibitor Cocktail, Roche) and phosphatase inhibitors (phosphatase inhibitor

cocktail 2, Sigma). Protein concentration was assessed using a Pierce BCA protein

assay kit (Thermo Fisher Scientific) according to manufacturer’s instructions. Proteins

were separated by SDS-PAGE under reducing conditions and transferred onto a

polyvinylidene fluoride membrane (0.45 μm, Amersham). The membrane was blocked

in 5% milk in PBS-T0.1 (Phosphate Buffer Saline, Tween 0.1%) for 1h and incubated

overnight at 4˚C with the primary antibodies anti-pAMPK (Abcam 2535S, dilution

1/1000), pRaptor (Abcam 2083S, dilution 1/1000), pACC (Abcam 3661S, dilution

1/1000), AMPK (Abcam 2532S, dilution 1/1000), Raptor (Abcam 2280S, dilution

1/1000), ACC (Abcam 3662S, dilution 1/1000), ANKRD23 (Abcam ab118210, dilution

(26)

1/500). The expression of α-tubulin (dilution 1/20000; T5168, Sigma) was used as a

loading control. After washing, the membrane was incubated for 1h in horseradish

peroxidase coupled secondary antibodies (Goat Anti-Rabbit-HRP, Abcam, AB6721,

dilution 1/8000) and the signal was detected on Amersham Hyperfilm ECL, after

exposure to Amersham ECL WB Detection Reagent. Band intensity was measured

using ImageJ.

8c. The role of Sec61G

The role of Sec61G was evaluated in INS-1E and EndoC-βH1 cells, cultured as

described [73–75] and further validated in dispersed human islets. Cells were

transfected overnight with 30 nM control siRNA (Qiagen) or siRNAs targeting rat or

human Sec61G as previously described [76]. Messenger RNA was isolated using the

Dynabeads mRNA DIRECT kit (Invitrogen, Paisley, UK) and reverse transcribed [77].

qRT-PCR was done using iQ SYBR Green Supermix (BIO-RAD, Nazareth Eke,

Belgium) on a LightCycler (Roche Diagnostics, Mannheim, Germany) or iCycler MyiQ

Single Color (BIORAD). Data were expressed as number of copies generated from the

standard curve method and corrected for the reference gene β-actin or

glyceraldehyde-3phosphate dehydrogenase (GAPDH). Primers used for qRT-PCR are

listed in the table below. Cells were exposed to 0.5 mM palmitate pre-complexed to

0.67% FFA-free BSA (16h for INS-1E cells, 24h for human islet cells) or to thapsigargin

for 24h. Apoptosis was assessed by fluorescence microscopy in propidium iodide (5

mg/ml) and Hoechst 33342 (5 mg/ml) stained cells as described [48]. For Western

blotting, INS-1E cells were lysed in Laemmli buffer and immunoblotted with cleaved

caspase 3 (Asp175, Cell Signaling Cat# 9661, dilution 1/1000), cleaved caspase 9

(Asp353, Cell Signaling Cat# 9507, 1/1000), and tubulin (Sigma Cat# T9026, 1/5000)

antibodies. Membranes were subsequently exposed to secondary

peroxidase-conjugated antibody (1/5000), and developed using SuperSignal West Femto

chemiluminescent substrate (Thermo Scientific) in a Bio-Rad chemi DocTM XRS +

(Bio-Rad laboratories).

(27)

Primers for rat and human GAPDH, ACTB, SEC61G

9. Expression quantitative trait loci (eQTL)

Specific details of the study were recently published [78]. Briefly, an eQTL analysis

was performed in a total of 100 islets from ND organ donor subjects. Genotyping was

performed using the 2.5 M Omniarray Beadchip and RNA analysis was performed

using Human Genome U133 Plus 2.0 Array to generate the eQTL analysis. Gender

and age were used as covariated and a cis-window of 500 kb was used. A nominal

p-value of p<0.05 was considered significant.

10. Statistical analysis

The results from the lipoglucotoxic conditions functional experiments are presented as

mean±SE and were analyzed with the R software or the GraphPad Prism Software

v8.4.2. Comparisons between two sets of data were performed by the two-tailed

Student’s t-test. Results from more than two groups were analyzed by ANOVA followed

by the Turkey correction. Multiple linear regression was used to take in to account the

effects of incubation time and culture conditions on multiple groups of islets. A p-value

< 0.05 was considered statistically significant.

Application Forward (5´-3´) Reverse (5´-3´) Species Length

GAPDH ST Standard curve ATGACTCTACCCACGGCAAG TGTGAGGGAGATGCTCAGTG Rat 930 bp

ACTB ST Standard curve AAATCTGGCACCACACCTTC CCGATCCACACGGAGTACTT Human 805 bp

SEC61G ST Standard curve TGGATCAGGTAATGCAGTTTG TAGGGATGTGGATCAGTTTCA Rat 180bp

SEC61G ST Standard curve TGGATCAGGTAATGCAGTTTG CAGCCACCAACAATGATGTT Human 205 bp

GAPDH RT Real time AGTTCAACGGCACAGTCAAG TACTCAGCACCAGCATCACC Rat 136 bp

ACTB RT Real time CTGTACGCCAACACAGTGCT GCTCAGGAGGAGCAATGATC Human 127 bp

SEC61G RT Real time AGT CGG CAG TTT GTA AAG GA GAA CCC CAT GAT AGC GAA T Rat 117 bp

(28)

RESULTS

1. Impact of different lipoglucotoxic conditions on ND islet beta-cell function

Glucose-stimulated insulin secretion (GSIS) of human islets under control or

lipoglucotoxic conditions was evaluated as schematically represented in Figure 1. It

was found that the impact of metabolic stress and washout differed according to the

experimental settings. In the islets cultured at 11.1 mmol/l glucose (g), 1.0 mmol/l

palmitate+oleate, (1:2 molar ratio, P+O) or 1.0 mmol/l palmitate+oleate+11.1 mmol/l

glucose (P+O+g), no significant change in GSIS occurred at D4 as compared to islets

cultured in control medium (5.5 mmol/l glucose), with similar GSIS at day 2 (D2; basal),

D4 (stress), and D8 (after washout) (Figure 3A-C); accordingly no modification was

observed in insulin stimulation index (ISI) (Figure 3D-F). The number of separate islet

preparations and replicates per condition were 4 and 7/8 per time point (38 replicates

in total) for g, 3 and 4/6 per time point (26 replicates in total) for P+O, 3 and 5/6 per

time point (28 replicates in total) for P+O+g.

A different scenario was seen with the islets cultured at 0.5 mmol/l palmitate (P), 22.2

mmol/l glucose (G), 0.5 mmol/l palmitate+11.1 mmol/l glucose (P+g), 0.5 mmol/l

palmitate+22.2 mmol/l glucose (P+G) or 1.0 mmol/l palmitate+oleate+22.2 mmol/l

glucose (P+O+G) which showed, at D4, several changes of GSIS (Figure 4). In detail,

0.5 mmol/l palmitate (P and P+g) induced a trend to lower insulin release in response

to acute stimulation with high (16.7 mmol/l) glucose by a -49.2% and -27.7%

respectively (Figure 4A, C). Two-day incubation with G was associated with a

tendency to higher at low (3.3 mmol/l) glucose (+49.6%) and reduced release in

response to 16.7 mmol/l glucose (Figure 4B). Exposure to the combination of palmitate

and high glucose (P+G and P+O+G) induced a +94.2% and +64.8% insulin secretion

increment at low glucose, respectively, but a less affected response to increased

glucose level (Figure 4D, E). These alterations reflected the significant reduction of

the respective ISI values at D4 in all five conditions, in comparison with the control at

the same time point (Figure 4F-J).

However, GSIS improved after washout of P, G and P+g, so that no significant

differences persisted at D8 between control and previously treated islets (Figure

4A-C, F-H). On the contrary, in the islets exposed to the lipoglucotoxic conditions P+G or

P+O+G the secretory dysfunction persisted even after the washout (D8) (Figure

(29)

4D,E,I,J). The number of separate islet preparations and replicates per condition were

4 and 7/8 per time point (37 replicates in total) for P, 4 and 7/8 per time point (37

replicates in total) for G, 3 and 5/6 per time point (29 replicates in total) for P+g, 3 and

5/6 per time point (28 replicates in total) for P+G, 6 and 10/12 per time point (54

replicates in total) for P+O+G.

Over the 8-day incubation time, islet insulin content significantly decreased (Figure 5).

In control islets, the insulin content tended to be lower at D4 (about -15%) and was

significantly reduced at D8 vs D2 (approximately -30%, p<0.01) (Figure 5a). Insulin

content was not available for the islets g-exposed series. For the islets incubated with

P+O and P+O+g (not associated with any change of insulin release), reduction of

insulin content resembled that of control islets (Figure 5b). For the culture conditions

causing dysfunction (P, G, P+g, P+G, and P+O+G), a markedly lower insulin content

(<50%) was already seen at D4, which remained reduced at D8 (Figure 5c, d). The

number of replicates for insulin content was 28 at each time point for control islets,

ranged from 7 to 12 at separate time points for the settings associated with no damage,

it was 15-16 at the time points for the conditions causing a functional damage at D4

with recovery at D8, and was 10-11 at separate time points for the conditions where

the damage persisted even after D8 washout. To confirm the functional tendency

observed throughout the time culture, actuaI ISI values were normalized to the

respective islet insulin content at each time point, showing similar results (Figure 6,

Figure 4F-J). As for the normalized ISI values, the number of separate islet

preparations and replicates were 3 and 3/6 per time point (46 replicates in total) for

P+O, 3 and 3/6 per time point (44 replicates in total) for P+O+g, 4 and 4 per time point

(40 replicates in total) for P, 3 and 5/6 per time point (58 replicates in total) for G, 3 and

5/6 per time point (54 replicates in total) for P+g, 3 and 4/6 per time point (52 replicates

in total) for P+G, 5 and 4/6 per time point (52 replicates in total) for P+O+G.

Normalization of ISI values for the g series was not available.

In summary, acute GSIS from human islets was impaired after prolonged exposure to

some, but not all of the tested lipoglucotoxic stressors, depending on the type and

concentration and their combination. Specifically, palmitate or high glucose induced a

damage that was reversible after their removal from the culture medium, when tested

alone; beta-cell dysfunction persisted when the fatty acid and high glucose were

combined.

Riferimenti

Documenti correlati

This long-delay design enabled us to clearly distinguish activation related to visual stimulation/encod- ing, memory maintenance during the delay period, and memory retrieval at

Even if the scores are quite low (we are facing a hard problem with a straightforward classifier), the results lead to some evident conclusions: 1) using solely the Google Images

This study investigates how certain organisational, managerial, and technological factors enable the adoption of software as a service (SaaS) model for customer

If also in this field precision medicine has significantly contributed to improving disease prognosis [4], three successive coronavirus epidemics (SARS, MERS

Several studies have shown that acute administration of testosterone induces a rapid relaxation in vascular tissues of different species including humans [8–11] , suggesting

1) Tutti quei casi in cui non può sottrarsi all’applicazione del diritto internazionale in quanto esso regola il rapporto dedotto in giudizio. Qui il giudice viene

Primary sanctions prohibit individuals and companies in the sanctioning State from carrying out business with the sanctioned country. These measures cause the interruption of

The GMRT radio observations at 610 MHz confirm the presence of a radio halo, which remarkably overlaps the X-ray cluster emission and connects a couple of radio relics.. We detect