Heparanase overexpression induces glucagon resistance and. protects animals from chemically-induced diabetes

Σχετικά έγγραφα
Cellular Physiology and Biochemistry

IL - 13 /IL - 18 ELISA PCR RT - PCR. IL - 13 IL - 18 mrna. 13 IL - 18 mrna IL - 13 /IL Th1 /Th2

ΠΡΟΓΡΑΜΜΑ ΜΕΤΑΠΤΥΧΙΑΚΩΝ ΣΠΟΥΔΩΝ ΣΤΙΣ «ΚΛΙΝΙΚΕΣ ΚΑΙ ΚΛΙΝΙΚΟΕΡΓΑΣΤΗΡΙΑΚΕΣ ΙΑΤΡΙΚΕΣ ΕΙΔΙΚΟΤΗΤΕΣ»

ΤΕΧΝΟΛΟΓΙΚΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΚΥΠΡΟΥ ΤΜΗΜΑ ΝΟΣΗΛΕΥΤΙΚΗΣ

Assalamu `alaikum wr. wb.

«ΑΓΡΟΤΟΥΡΙΣΜΟΣ ΚΑΙ ΤΟΠΙΚΗ ΑΝΑΠΤΥΞΗ: Ο ΡΟΛΟΣ ΤΩΝ ΝΕΩΝ ΤΕΧΝΟΛΟΓΙΩΝ ΣΤΗΝ ΠΡΟΩΘΗΣΗ ΤΩΝ ΓΥΝΑΙΚΕΙΩΝ ΣΥΝΕΤΑΙΡΙΣΜΩΝ»

Math 6 SL Probability Distributions Practice Test Mark Scheme

CHAPTER 25 SOLVING EQUATIONS BY ITERATIVE METHODS

ΤΕΧΝΟΛΟΓΙΚΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΚΥΠΡΟΥ ΣΧΟΛΗ ΓΕΩΠΟΝΙΚΩΝ ΕΠΙΣΤΗΜΩΝ ΒΙΟΤΕΧΝΟΛΟΓΙΑΣ ΚΑΙ ΕΠΙΣΤΗΜΗΣ ΤΡΟΦΙΜΩΝ. Πτυχιακή εργασία

ΠΑΝΕΠΙΣΤΗΜΙΟ ΠΕΙΡΑΙΑ ΤΜΗΜΑ ΝΑΥΤΙΛΙΑΚΩΝ ΣΠΟΥΔΩΝ ΠΡΟΓΡΑΜΜΑ ΜΕΤΑΠΤΥΧΙΑΚΩΝ ΣΠΟΥΔΩΝ ΣΤΗΝ ΝΑΥΤΙΛΙΑ

2 Composition. Invertible Mappings

ΕΠΙΔΡΑΣΗ ΤΗΣ ΧΡΗΣΗΣ ΕΛΑΙΟΠΛΑΚΟΥΝΤΑ ΣΤΗΝ ΔΙΑΤΡΟΦΗ ΤΩΝ ΑΙΓΩΝ ΔΑΜΑΣΚΟΥ ΩΣ ΠΡΟΣ ΤΗΝ ΠΟΣΟΤΗΤΑ ΚΑΙ ΠΟΙΟΤΗΤΑ ΤΟΥ ΠΑΡΑΓΟΜΕΝΟΥ ΓΑΛΑΚΤΟΣ

Strain gauge and rosettes

Cinnamaldehyde Prevents Endothelial Dysfunction Induced by High Glucose by Activating Nrf2

ΙΠΛΩΜΑΤΙΚΗ ΕΡΓΑΣΙΑ. ΘΕΜΑ: «ιερεύνηση της σχέσης µεταξύ φωνηµικής επίγνωσης και ορθογραφικής δεξιότητας σε παιδιά προσχολικής ηλικίας»

«Συντήρηση αχλαδιών σε νερό. υπό την παρουσία σπόρων σιναπιού (Sinapis arvensis).»

Electrolyzed-Reduced Water as Artificial Hot Spring Water

EE512: Error Control Coding

ΠΑΝΔΠΗΣΖΜΗΟ ΠΑΣΡΩΝ ΣΜΖΜΑ ΖΛΔΚΣΡΟΛΟΓΩΝ ΜΖΥΑΝΗΚΩΝ ΚΑΗ ΣΔΥΝΟΛΟΓΗΑ ΤΠΟΛΟΓΗΣΩΝ ΣΟΜΔΑ ΤΣΖΜΑΣΩΝ ΖΛΔΚΣΡΗΚΖ ΔΝΔΡΓΔΗΑ

the total number of electrons passing through the lamp.

Μελέτη της έκφρασης του ογκοκατασταλτικού γονιδίου Cyld στον καρκίνο του μαστού

ΑΡΙΣΤΟΤΕΛΕΙΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΘΕΣΣΑΛΟΝΙΚΗΣ ΤΜΗΜΑ ΟΔΟΝΤΙΑΤΡΙΚΗΣ ΕΡΓΑΣΤΗΡΙΟ ΟΔΟΝΤΙΚΗΣ ΚΑΙ ΑΝΩΤΕΡΑΣ ΠΡΟΣΘΕΤΙΚΗΣ

HOMEWORK 4 = G. In order to plot the stress versus the stretch we define a normalized stretch:

Group 2 Methotrexate 7.5 mg/week, increased to 15 mg/week after 4 weeks. Methotrexate 7.5 mg/week, increased to 15 mg/week after 4 weeks

ΜΕΤΑΠΤΥΧΙΑΚΗ ΕΡΕΥΝΗΤΙΚΗ ΔΙΑΤΡΙΒΗ

[1] P Q. Fig. 3.1

High mobility group 1 HMG1

Longitudinal Changes in Component Processes of Working Memory

; +302 ; +313; +320,.

Μαρία Κατσιφοδήμου. Ο ρόλος της έκκρισης HLA-G από τα ανθρώπινα έμβρυα στην επιτυχία της εξωσωματικής γονιμοποίησης. Μεταπτυχιακή Διπλωματική Εργασία

Other Test Constructions: Likelihood Ratio & Bayes Tests

Αξιοποίηση Φυσικών Αντιοξειδωτικών στην Εκτροφή των Αγροτικών

Mean bond enthalpy Standard enthalpy of formation Bond N H N N N N H O O O

Πτυχιακή Εργασία Η ΠΟΙΟΤΗΤΑ ΖΩΗΣ ΤΩΝ ΑΣΘΕΝΩΝ ΜΕ ΣΤΗΘΑΓΧΗ

ΠΕΡΙΕΧΟΜΕΝΑ. Κεφάλαιο 1: Κεφάλαιο 2: Κεφάλαιο 3:

Optimizing Microwave-assisted Extraction Process for Paprika Red Pigments Using Response Surface Methodology

ΘΕΩΡΗΤΙΚΗ ΚΑΙ ΠΕΙΡΑΜΑΤΙΚΗ ΙΕΡΕΥΝΗΣΗ ΤΗΣ ΙΕΡΓΑΣΙΑΣ ΣΚΛΗΡΥΝΣΗΣ ΙΑ ΛΕΙΑΝΣΕΩΣ

Démographie spatiale/spatial Demography

ΚΥΠΡΙΑΚΗ ΕΤΑΙΡΕΙΑ ΠΛΗΡΟΦΟΡΙΚΗΣ CYPRUS COMPUTER SOCIETY ΠΑΓΚΥΠΡΙΟΣ ΜΑΘΗΤΙΚΟΣ ΔΙΑΓΩΝΙΣΜΟΣ ΠΛΗΡΟΦΟΡΙΚΗΣ 19/5/2007

ΤΕΧΝΟΛΟΓΙΚΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΚΥΠΡΟΥ ΣΧΟΛΗ ΕΠΙΣΤΗΜΩΝ ΥΓΕΙΑΣ ΤΜΗΜΑ ΝΟΣΗΛΕΥΤΙΚΗΣ ΠΤΥΧΙΑΚΗ ΕΡΓΑΣΙΑ ΕΠΗΡΕΑΖΕΙ ΤΗΝ ΠΡΟΛΗΨΗ ΚΑΡΚΙΝΟΥ ΤΟΥ ΜΑΣΤΟΥ

ΑΡΙΣΤΟΤΕΛΕΙΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΘΕΣΣΑΛΟΝΙΚΗΣ ΙΑΤΡΙΚΗ ΣΧΟΛΗ. ΠΡΟΓΡΑΜΜΑ ΜΕΤΑΠΤΥΧΙΑΚΩΝ ΣΠΟΥ ΩΝ «Ιατρική Ερευνητική Μεθοδολογία» ΙΠΛΩΜΑΤΙΚΗ ΕΡΓΑΣΙΑ

ΠΤΥΧΙΑΚΗ ΕΡΓΑΣΙΑ "ΠΟΛΥΚΡΙΤΗΡΙΑ ΣΥΣΤΗΜΑΤΑ ΛΗΨΗΣ ΑΠΟΦΑΣΕΩΝ. Η ΠΕΡΙΠΤΩΣΗ ΤΗΣ ΕΠΙΛΟΓΗΣ ΑΣΦΑΛΙΣΤΗΡΙΟΥ ΣΥΜΒΟΛΑΙΟΥ ΥΓΕΙΑΣ "

Παρουσίαση ερευνητικού έργου

Capacitors - Capacitance, Charge and Potential Difference

ACTA CHINESE MEDICINE. diabetic nephropathies DN 24. urine protein quantitation in 24 hours 24hUTP serum creatinine Scr

ΠΑΝΕΠΙΣΤΗΜΙΟ ΠΑΤΡΩΝ ΣΧΟΛΗ ΑΝΘΡΩΠΙΣΤΙΚΩΝ ΚΑΙ ΚΟΙΝΩΝΙΚΩΝ ΕΠΙΣΤΗΜΩΝ ΤΜΗΜΑ ΦΙΛΟΛΟΓΙΑΣ

Contents Part I Psychoneuroimmunology and Systems Biology Mechanisms 1 From Psychoneuroimmunology to Personalized, Systems, and Dynamical Medicine

ΤΕΧΝΟΛΟΓΙΚΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΚΥΠΡΟΥ ΣΧΟΛΗ ΓΕΩΤΕΧΝΙΚΩΝ ΕΠΙΣΤΗΜΩΝ ΚΑΙ ΔΙΑΧΕΙΡΙΣΗΣ ΠΕΡΙΒΑΛΛΟΝΤΟΣ. Πτυχιακή εργασία

Table S1: Inpatient Diet Composition

ΤΕΧΝΟΛΟΓΙΚΟ ΕΚΠΑΙ ΕΥΤΙΚΟ Ι ΡΥΜΑ ΚΡΗΤΗΣ ΣΧΟΛΗ ΙΟΙΚΗΣΗΣ ΚΑΙ ΟΙΚΟΝΟΜΙΑΣ ΤΜΗΜΑ ΙΟΙΚΗΣΗΣ ΕΠΙΧΕΙΡΗΣΕΩΝ ΠΤΥΧΙΑΚΗ ΕΡΓΑΣΙΑ

ΓΕΩΠΟΝΙΚΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΑΘΗΝΩΝ ΤΜΗΜΑ ΑΓΡΟΤΙΚΗΣ ΟΙΚΟΝΟΜΙΑΣ & ΑΝΑΠΤΥΞΗΣ

Μηχανισμοί πρόβλεψης προσήμων σε προσημασμένα μοντέλα κοινωνικών δικτύων ΔΙΠΛΩΜΑΤΙΚΗ ΕΡΓΑΣΙΑ

[11].,, , 316 6, ,., 15.5%, 9.8%, 2006., IDF,, ,500, 2,830.,, ,200.,,, β, [12]. 90% 2,,,,, [13-15].,, [13,

ΕΘΝΙΚΗ ΣΧΟΛΗ ΔΗΜΟΣΙΑΣ ΔΙΟΙΚΗΣΗΣ ΙΓ' ΕΚΠΑΙΔΕΥΤΙΚΗ ΣΕΙΡΑ

Physical and Chemical Properties of the Nest-site Beach of the Horseshoe Crab Rehabilitated by Sand Placement

HIV HIV HIV HIV AIDS 3 :.1 /-,**1 +332

Αξιοποίηση Φυσικών Αντιοξειδωτικών στην Εκτροφή των Αγροτικών

ΤΕΧΝΟΛΟΓΙΚΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΚΥΠΡΟΥ ΣΧΟΛΗ ΓΕΩΤΕΧΝΙΚΩΝ ΕΠΙΣΤΗΜΩΝ ΚΑΙ ΔΙΑΧΕΙΡΙΣΗΣ ΠΕΡΙΒΑΛΛΟΝΤΟΣ. Πτυχιακή εργασία

ΤΕΧΝΟΛΟΓΙΚΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΚΥΠΡΟΥ ΣΧΟΛΗ ΓΕΩΠΟΝΙΚΩΝ ΕΠΙΣΤΗΜΩΝ ΚΑΙ ΔΙΑΧΕΙΡΙΣΗΣ ΠΕΡΙΒΑΛΛΟΝΤΟΣ. Πτυχιακή διατριβή

Απόκριση σε Μοναδιαία Ωστική Δύναμη (Unit Impulse) Απόκριση σε Δυνάμεις Αυθαίρετα Μεταβαλλόμενες με το Χρόνο. Απόστολος Σ.

Congruence Classes of Invertible Matrices of Order 3 over F 2

Η ΕΠΙΔΡΑΣΗ ΤΗΣ ΑΙΘΑΝΟΛΗΣ,ΤΗΣ ΜΕΘΑΝΟΛΗΣ ΚΑΙ ΤΟΥ ΑΙΘΥΛΟΤΡΙΤΟΤΑΓΗ ΒΟΥΤΥΛΑΙΘΕΡΑ ΣΤΙΣ ΙΔΙΟΤΗΤΕΣ ΤΗΣ ΒΕΝΖΙΝΗΣ

Every set of first-order formulas is equivalent to an independent set

derivation of the Laplacian from rectangular to spherical coordinates

Emulsifying Properties of Egg Yolk as a Function of Diacylglycerol Oil

«Χρήσεις γης, αξίες γης και κυκλοφοριακές ρυθμίσεις στο Δήμο Χαλκιδέων. Η μεταξύ τους σχέση και εξέλιξη.»

Potential Dividers. 46 minutes. 46 marks. Page 1 of 11

Μηχανική Μάθηση Hypothesis Testing

Η ΥΠΕΡΕΚΦΡΑΣΗ ΤΟΥ SMAD7 ΠΡΟΣΤΑΤΕΥΕΙ ΤΟ ΗΠΑΡ ΑΠΟ ΤΗΝ TGF-Β/SMAD ΜΕΣΟΛΑΒΟΥΜΕΝΗ ΙΝΟΓΕΝΕΣΗ

Main source: "Discrete-time systems and computer control" by Α. ΣΚΟΔΡΑΣ ΨΗΦΙΑΚΟΣ ΕΛΕΓΧΟΣ ΔΙΑΛΕΞΗ 4 ΔΙΑΦΑΝΕΙΑ 1

ΕΙΣΑΓΩΓΗ ΣΤΗ ΣΤΑΤΙΣΤΙΚΗ ΑΝΑΛΥΣΗ

Αζεκίλα Α. Μπνπράγηεξ (Α.Μ. 261)

(1) Describe the process by which mercury atoms become excited in a fluorescent tube (3)

þÿ Ç»¹º ³µÃ ± : Ãż²» Ä Â

C.S. 430 Assignment 6, Sample Solutions

ST5224: Advanced Statistical Theory II

Second Order RLC Filters

5.4 The Poisson Distribution.

Comparison of Evapotranspiration between Indigenous Vegetation and Invading Vegetation in a Bog

Reaction of a Platinum Electrode for the Measurement of Redox Potential of Paddy Soil

Study of In-vehicle Sound Field Creation by Simultaneous Equation Method

ΓΕΩΠΟΝΙΚΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΑΘΗΝΩΝ ΤΜΗΜΑ ΕΠΙΣΤΗΜΗΣ ΤΡΟΦΙΜΩΝ ΚΑΙ ΔΙΑΤΡΟΦΗΣ ΤΟΥ ΑΝΘΡΩΠΟΥ

ΤΕΧΝΟΛΟΓΙΚΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΚΥΠΡΟΥ ΣΧΟΛΗ ΕΠΙΣΤΗΜΩΝ ΥΓΕΙΑΣ

Does anemia contribute to end-organ dysfunction in ICU patients Statistical Analysis

ΕΦΑΡΜΟΓΗ ΕΥΤΕΡΟΒΑΘΜΙΑ ΕΠΕΞΕΡΓΑΣΜΕΝΩΝ ΥΓΡΩΝ ΑΠΟΒΛΗΤΩΝ ΣΕ ΦΥΣΙΚΑ ΣΥΣΤΗΜΑΤΑ ΚΛΙΝΗΣ ΚΑΛΑΜΙΩΝ

ΓΕΩΠΟΝΙΚΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΑΘΗΝΩΝ ΤΜΗΜΑ ΑΓΡΟΤΙΚΗΣ ΟΙΚΟΝΟΜΙΑΣ & ΑΝΑΠΤΥΞΗΣ

Διδακτορική Διατριβή

ΤΕΧΝΟΛΟΓΙΚΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΚΥΠΡΟΥ ΤΜΗΜΑ ΝΟΣΗΛΕΥΤΙΚΗΣ

ΠΑΡΑΜΕΤΡΟΙ ΕΠΗΡΕΑΣΜΟΥ ΤΗΣ ΑΝΑΓΝΩΣΗΣ- ΑΠΟΚΩΔΙΚΟΠΟΙΗΣΗΣ ΤΗΣ BRAILLE ΑΠΟ ΑΤΟΜΑ ΜΕ ΤΥΦΛΩΣΗ

Section 8.3 Trigonometric Equations

Resurvey of Possible Seismic Fissures in the Old-Edo River in Tokyo

Επιβλέπουσα Καθηγήτρια: ΣΟΦΙΑ ΑΡΑΒΟΥ ΠΑΠΑΔΑΤΟΥ

The challenges of non-stable predicates

Repeated measures Επαναληπτικές μετρήσεις

ΑΡΙΣΤΟΤΕΛΕΙΟ ΠΑΝΕΠΙΣΤΗΜΙΟ ΘΕΣΣΑΛΟΝΙΚΗΣ ΔΙΑΤΡΟΦΙΚΕΣ ΣΥΝΗΘΕΙΕΣ, ΚΟΙΝΩΝΙΚΑ & ΑΝΘΡΩΠΟΜΕΤΡΙΚΑ ΧΑΡΑΚΤΗΡΙΣΤΙΚΑ ΕΛΛΗΝΩΝ ΕΠΑΓΓΕΛΜΑΤΙΩΝ ΠΟΔΟΣΦΑΡΙΣΤΩΝ

*,* + -+ on Bedrock Bath. Hideyuki O, Shoichi O, Takao O, Kumiko Y, Yoshinao K and Tsuneaki G

Φυσικοθεραπευτής, MSc, Εργαστηριακός συνεργάτης, Τμήμα Φυσικοθεραπείας, ΑΤΕΙ Λαμίας Φυσικοθεραπευτής

Aluminum Electrolytic Capacitors

Γεωπονικό Πανεπιςτήμιο Αθηνών Τμήμα Αξιοποίηςησ Φυςικών Πόρων και Γεωργικήσ Μηχανικήσ

ΙΔΡΥΜΑ. Θεσσαλονίκη, ύλα

Transcript:

Page 1 of 85 Heparanase overexpression induces glucagon resistance and protects animals from chemically-induced diabetes Dahai Zhang 1, Fulong Wang 1, Nathaniel Lal 1, Amy Pei-Ling Chiu 1, Andrea Wan 1, Jocelyn Jia 1, Denise Bierende 1, Stephane Flibotte 1, Sunita Sinha 1, Ali Asadi 2, Xiaoke Hu 2, Farnaz Taghizadeh 2, Thomas Pulinilkunnil 3, Corey Nislow 1, Israel Vlodavsky 4, James D. Johnson 2, Timothy J. Kieffer 2, Bahira Hussein 1 and Brian Rodrigues 1 1 Faculty of Pharmaceutical Sciences, UBC, 2405 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3; 2 Department of Cellular & Physiological Sciences, Life Sciences Institute, UBC, 2350 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3; 3 Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, 100 Tucker Park Road, Saint John, NB, Canada E2L 4L5; 4 Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel 31096 Running Title: Heparanase overexpression and the pancreatic islet Corresponding author: Dr. Brian Rodrigues Faculty of Pharmaceutical Sciences University of British Columbia, 2405 Wesbrook Mall, Vancouver, B.C., Canada V6T 1Z3 TEL: (604) 822-4758; FAX: (604) 822-3035 E-mail: rodrigue@mail.ubc.ca Key Words: Heparanase, heparan sulfate proteoglycan, glucose homeostasis, glucagon resistance, pancreatic islet, STZ Word Count: 4761 Total Number of Figures: Publish 6 Ahead of Print, published online October 7, 2016

Page 2 of 85 Abstract Heparanase, a protein with enzymatic and non-enzymatic properties, contributes towards disease progression and prevention. In the current study, a fortuitous observation in transgenic mice globally overexpressing heparanase (hep-tg) was the discovery of improved glucose homeostasis. We examined the mechanisms that contribute towards this improved glucose metabolism. Heparanase overexpression was associated with enhanced GSIS and hyperglucagonemia, in addition to changes in islet composition and structure. Strikingly, the pancreatic islet transcriptome was greatly altered in hep-tg mice with over 2000 genes differentially expressed versus control. The upregulated genes were enriched for diverse functions including cell death regulation, extracellular matrix component synthesis, and pancreatic hormone production. The downregulated genes were tightly linked to regulation of the cell cycle. In response to multiple low-dose STZ, hep-tg animals developed less severe hyperglycemia compared to WT, an effect likely related to their beta cells being more functionally efficient. In animals given a single, high dose of STZ, causing severe and rapid development of hyperglycemia related to the catastrophic loss of insulin, hep-tg mice continued to have significantly lower blood glucose. In these mice, protective pathways were uncovered for managing hyperglycemia and include augmentation of FGF21 and GLP-1. This study uncovers the opportunity to utilize properties of heparanase in management of diabetes.

Page 3 of 85 Introduction Heparan sulfate proteoglycans (HSPG), located mainly on the cell surface and in the extracellular matrix, are composed of a core protein to which one or more heparan sulfate (HS) side chains are attached (1). HSPGs function, not only as structural proteins, but also as anchors for bioactive molecules, as HS is negatively charged. Highly expressed in pancreatic islets, HS binds and guides the signalling and distribution of fibroblast growth factor (FGF) family members, which regulate pancreatic endocrine cell differentiation, clustering and development (2). It has been suggested that the presence of HSPG in the nucleus has a suppressive effect on histone acetyltransferase activity and may therefore modulate gene expression (3). Heparanase is an endo-β-d-glucuronidase that is ubiquitously expressed in many organs, with blood and endothelial cells having the highest expression. Heparanase is encoded as a 65-kDa latent precursor (Hep L ) that requires proteolytic cleavage to form an active enzyme (Hep A ) (4). Functionally, Hep A cleaves HS at D-glucuronic acid residues, an action associated with extracellular matrix disruption (5) and release of cell surface-bound molecules such as FGF (2). Aside from the function of Hep A in cleaving HS, Hep L can also activate numerous signalling elements including Erk1/2, PI3K-AKT, RhoA and Src (6-9). These signalling events, in addition to the entry of Hep A into the nucleus to regulate histone acetylation by cleaving HS, have been suggested as mechanisms for modulating gene expression (3). Heparanase has physiological functions in wound healing and hair growth (10). However, intensive research has also hinted towards an additional role of heparanase in both disease progression and prevention. Thus, in cancer, degradation of HS chains by the increased expression of heparanase is associated with extracellular matrix and basement membrane (BM) disruption; loss of this physical barrier facilitates tumour cell invasion (11). In the pancreas, islet-enriched HS blocks immune cell infiltration, reduces T-cell mediated

and protection from streptozotocin (STZ)-induced diabetes. Page 4 of 85 inflammation (12), and supports beta cell survival and function (13). Thus, removal of HS by heparanase is critical for the initiation and progression of autoimmune (Type 1) diabetes (12). Additionally, heparanase expression is induced in many organs during diabetes. In the heart, such inappropriate expression may amplify fatty acid delivery and utilization. These events, if sustained, can lead to lipotoxicity and cardiovascular disease (14). In contrast to these observations, heparanase has also been shown to have beneficial effects in some diseases. For instance, overexpression of this protein is protective against adriamycin-induced kidney injury (15). In the brain, fragmentation of HS as a result of heparanase cleavage reduced amyloid deposition in Alzheimer s disease (16). Similarly, amyloid formation in the islets, a hallmark of Type 2 diabetes, could be ameliorated by heparanase-induced matrix metalloproteinase-9 (MMP-9) expression (17; 18). MMP-9 constitutes an endogenous islet protease that limits islet amyloid deposition and its concomitant toxic effects via degradation of the amyloid polypeptide (19). Thus, precise temporal and spatial control of heparanase may be essential for normal cell physiology. In the current study, when monitoring the metabolism of transgenic mice that globally overexpress heparanase (hep-tg), we made the fortuitous observation that glucose homeostasis was improved in these animals. Compared to wild type (WT) controls, these mice had lower plasma insulin and blood glucose levels; however, they exhibited higher glucagon concentrations, suggesting the presence of glucagon resistance. Given that glucagon receptor knockout (GRKO) mice exhibited increased insulin sensitivity and resistance to diabetes (20; 21), we followed up these preliminary observations by examining whether heparanase overexpression could contribute towards improving glucose metabolism as well as resistance to chemically-induced diabetes. Our data indicate that heparanase overexpression is associated with dramatic shifts in hormones secreted from the pancreas, reorganization of islet composition and structure, significant changes in islet gene expression,

Page 5 of 85 Research Design and Methods Experimental Animals Wild type (WT) C57BL/6J mice aged 10 to 12 weeks were purchased from Charles River Laboratories. Heparanase transgenic mice (hep-tg), in which a constitutive β-actin promoter drives the expression of human heparanase gene in a C57BL/6J genetic background, were a kind gift from Dr. Israel Vlodavsky. All animals were housed in 12-h light/dark cycle, and pathogen-free conditions. Hep-tg mice were previously crossed for 10 generations with C57BL/6J mice to produce a stable homozygous background. Male homozygous hep-tg mice aged 12 to 15 weeks were used for all experiments. For confirmation of genotype, genomic DNA was prepared from 21 day-old animal ear punched tissue, and analyzed by PCR (Fig. 1 A) as previously detailed (10). All experiments were approved by the University of British Columbia Animal Care Committee, and were performed in accordance with the Canadian Council on Animal Care Guidelines. Metabolic Assessments and Treatment of Animals For measurement of basal blood glucose and plasma hormones, animals were fasted for 6 or 16 h and blood sampled from the tail vein. Blood glucose was measured using an ACCU- CHEK glucose monitor (Roche, Basel, Switzerland). Blood was also collected using a heparinized micro-tube (Fisher Scientific, Waltham, MA, USA), and centrifuged immediately for separation of plasma. Circulating hormones in the plasma were measured using the following kits: mouse insulin (ALPCO, Salem, NH, USA), glucagon and GLP-1 (Meso Scale Discovery, Rockville, MD, USA), FGF21 (R&D Systems, Minneapolis, MN, USA). For the oral glucose tolerance test (OGTT) or intraperitoneal (i.p.) glucose tolerance test (IPGTT), animals were fasted overnight (16 h), 2 g/kg glucose administered orally or i.p., and blood glucose measured at the indicated times. A different cohort of animals was treated in the same manner for the assessment of insulin following the OGTT. To perform the glucagon challenge,

Page 6 of 85 glucagon (1 µg/kg body weight) was administered i.p. into 6 or 16 h fasted mice, and blood glucose measured at the indicated times. The insulin (0.75 U/kg body weight) and L-arginine (2 g/kg, i.p.) challenge were performed the same way in 6 h fasted mice. In addition to glucose measurement following injection of L-arginine, plasma insulin and glucagon were also measured at the indicated times. For evaluation of skeletal muscle sensitivity to insulin, mice were fasted for 6 h prior to an i.p. injection of 2 µg/kg insulin. 10 min post insulin injection, skeletal muscle (soleus and gastrocnemius) was isolated for Western Blot determination of phospho- and total Akt (Cell Signalling, Danvers, MA, USA), with beta actin (Santa Cruz, Dallas, TX, USA) used as an internal control. Induction Streptozotocin (STZ) is a beta-cell selective toxin. Using STZ, we employed two different strategies to induce diabetes. With the first protocol, we injected multiple low doses of STZ (MLD-STZ, 50 mg/kg i.p.) for 5 consecutive days. This procedure induces gradual beta cell death, followed by an immune system response (22). Body weight and plasma glucose levels were monitored daily after the first STZ injection. Metabolic assessment and organ isolation were performed 1 week after the last STZ injection, following confirmation of hyperglycemia. We also used a single high dose of STZ (SHD-STZ, 200 mg/kg i.p.), which is directly cytotoxic to beta cells, resulting in robust hyperglycemia within 24-48 h (23). Staining and Quantification The pancreas from normal or MLD-STZ mice was perfused with PBS, harvested, fixed in 4% paraformaldehyde, and stored in 70% ethanol before paraffin embedding. All paraffin sections (5-µm thickness) were processed by Wax-it Histology laboratories (Vancouver, BC, Canada). Immunofluorescence staining and quantification of insulin (Cell Signalling Technology, Beverly, MA, USA), glucagon (Sigma-Aldrich, St. Louis, MO, USA), GLUT2 (Chemicon International Inc, Temecula, CA) and synaptophysin (Novus Biologicals, Littleton, CO, USA) were

Page 7 of 85 performed as previously described (24). Alcian blue (0.65 M MgCl2, ph 5.8; Sigma-Aldrich, St. Louis, MO, USA) staining and imaging was used to visualize HS. Western Blot Western blot was done as described previously (25). Heparanase (Abcam, Cambridge, UK) and heparan sulfate (Santa Cruz, Dallas, TX, USA) expression in the pancreas were determined using the appropriate antibodies. Islet Isolation Pancreatic islets were isolated using collagenase as described previously (13; 26). Briefly, the pancreas was perfused with collagenase through the pancreatic duct. Additional collagenase digestion was performed in a water bath. Following digestion, the tube containing the tissue sample was shaken vigorously. Individual islets were handpicked under a bright-field microscope. Islets were cultured for 3 h (37 C, 5% CO 2 ) in RPMI1640 medium (Invitrogen, Carlsbad, CA, USA) with 5 mm glucose (Sigma, St. Louis, MO, USA), 100 units/ml penicillin, 100 µg/ml streptomycin (Invitrogen), and 10% FBS (Invitrogen). For RNA sequencing, 100 islets were selected, rinsed with PBS and snap frozen for later isolation of RNA. For glucoseinduced insulin secretion, 10 islets were rinsed three times with PBS to remove glucose and insulin, and then transferred to RPMI1640 medium with either 3 mm or 16.7 mm glucose for 15 mins, followed by medium collection and insulin determination using ELISA. The total insulin content in islets was determined following acid-ethanol extraction. RNA Sequencing and Analysis RNA from 5 hep-tg and 5 WT mice was isolated using an RNeasy purification kit (Qiagen, Hilden, Germany). Sequencing libraries were prepared from 400 ng total RNA using the TruSeq Stranded mrna Sample Preparation kit according to the manufacturer s instructions (Illumina, San Diego, USA). Samples were checked for quality using a Bioanalyzer (Agilent,

Page 8 of 85 then multiplexed and sequenced over one rapid run lane on the HiSeq2500 (Illumina), collecting 89 million 100 bp paired-end reads. Kallisto version 0.42.4 was first used to build an index file for the mouse reference transcriptome GRCm38 as downloaded from the Ensembl web site (www.ensembl.org). The sequence reads for each sample were then quantified with the quant function of Kallisto. In house Perl scripts were used to sum the read counts at the transcript level for each gene and to create a matrix comprising the read counts for all the genes for all the samples. Differential expression analysis was then performed on the data from that matrix using the R package DESeq2 (27). Each sample was assessed using the quality control software RSeQC version 2.6.3 (28) and the PtR script from the trinity suite (29). One potential outlier was detected when clustering the samples and was therefore removed for the differential expression analysis. RNA sequencing transcriptomic data were analyzed using Panther and DAVID. Network analysis was conducted using STRING. Statistical Analysis Statistics were performed using Sigma Plot (Systat Software Inc., Chicago, IL, USA). For all analysis, student t-test or one-way ANOVA was used to determine differences between group mean values. Values are presented as means ± SEM with individual data points. p<0.05 was considered statistically significant.

Page 9 of 85 Results Heparanase Transgenic Mice Have Improved Glucose Homeostasis At 3 months of age, hep-tg mice had similar body weights compared to WT (hep-tg, 22.03±1.11 g; WT 22.11±1.08 g). However, the determination of circulating plasma insulin in hep-tg animals after a 6 h fast revealed lower levels compared to WT (Fig. 1B). Interestingly, the transgenic animals also demonstrated reduced basal blood glucose (Fig. 1C), suggesting higher insulin efficiency in these mice. This was tested by administering a bolus dose of insulin and measuring Akt activation in skeletal muscle. Samples of skeletal muscle from heptg mice showed an enhanced insulin response (Fig. 1D). To pursue this observation, we performed an OGTT after an overnight fast. Unlike in the case of an acute fast, prolonged fasting eliminated the difference in blood glucose between the two genotypes (Fig. 1E). In addition, although hep-tg mice tended to clear glucose faster, this improvement was not statistically significant (Fig. 1E), suggesting that the improvement in insulin sensitivity observed under basal conditions is not readily apparent during an OGTT. Following an insulin tolerance test, we were also unable to detect any difference in blood glucose lowering between the two groups, up to 30 min after insulin injection. Interestingly, after 30 min of insulin, we start to see a separation (recovery of glucose levels) between the WT and hep-tg mice (Supplementary Fig. 1). Unexpectedly, in response to the glucose challenge, insulin secretion in hep-tg animals was almost 2-fold higher than in WT (Fig. 1F). In addition, when islets from WT and hep-tg mice were isolated and exposed to high glucose, hep-tg islets were capable of releasing greater amounts of insulin compared to WT (Fig. 1G). Although hep-tg islets appeared to contain slightly more insulin compared to WT, this difference was not statistically significant (Supplementary Fig. 2). Similarly, as neither Ins 1 nor Ins 2 expression demonstrated any significant difference between the two strains (data not shown), our data

Page 10 of 85 suggest that heparanase overexpression influences the secretory process of insulin rather than its synthesis. Heparanase Overexpression Induces Glucagon Resistance And Changes In Pancreatic Islet Structure Intriguingly, hep-tg mice showed an augmentation in circulating basal glucagon levels (Fig. 2A and Supplementary Fig. 3). However, these mice demonstrated resistance when given a glucagon challenge after 6 (Supplementary Fig. 3) or 16 h (Fig. 2B) of fasting. The glucagon resistance in hep-tg mice was evident in the absence of any change in the glucagon receptor in the liver (Supplementary Fig. 4). In response to L-arginine, which drives islet hormone secretion independent of glucose sensing, hep-tg mice showed higher glucagon and lower insulin secretion, effects that were reflected in a higher glucose excursion in these animals (Fig. 2C). To determine the basis for this observed hyperglucagonemia, we quantified pancreatic alpha cells, and their ratio to beta cells, using fluorescent staining. Expressed as glucagon positive (WT, 0.000465 ± 0.00026; hep-tg, 0.000764 ± 0.000318; p=0.433) and insulin positive (WT, 0.004056 ± 0.000991; hep-tg, 0.004608 ± 0.001674; p=0.056) area individually (signal positive area versus total area of the section to represent the alpha and beta cell number in the pancreas), there was no statistical difference between WT and hep-tg animals in the number of insulin or glucagon positive cells. However, when the data was expressed as a ratio of alpha/beta cells, the hep-tg islets demonstrated alpha cells that were more abundant. Additionally, unlike WT islets, wherein alpha cells are mostly found in the periphery and beta cells in the core, hep-tg islets contained alpha cells that were randomly distributed (Fig. 2D). Islets in hep-tg mice exhibit similar average size and size distribution (Fig. 2E), and no difference in their number compared to WT (Supplementary Fig. 5). Overall, heparanase overexpression was associated with dramatic shifts in hormones secreted from the pancreas, in addition to changes in islet composition and structure.

Page 11 of 85 Heparanase Overexpression Greatly Alters The Pancreatic Islet Transcriptome Entry of heparanase into the nucleus to regulate histone acetylation has been put forth as a mechanism to modulate transcription (3). Given the impact of heparanase overexpression on insulin and glucagon, we sought to characterize the hep-tg islet transcriptome using RNAsequencing (Supplementary Table 1). As expected, heparanase was the most profoundly altered mrna in the islet transcriptome, exhibiting a >100 fold change (Fig. 3B). This was mirrored by an equally robust increase in heparanase protein expression (Fig. 3B, inset). Strikingly, after correction for multiple testing, over 2000 genes were significantly differentially expressed with p<0.05. Of these, 1176 were upregulated and 985 were downregulated (Fig. 3A, inset). From the mrnas that increased above a cut-off (>2-fold, p<0.001), we identified 350 genes enriched in multiple cellular processes including in development (e.g. Crlf1, Ifrd1) (30), metabolism (e.g. Acacb, Ppargc1a) (31), and cell death regulation (e.g., Npas4, Igf1, P2rx1) (Fig. 3A) (32; 33). Of note, glucagon (Gcg) expression increased (Fig. 3B), and corresponded to the high plasma glucagon observed in hep-tg mice. Among the 112 genes that decreased (>2-fold, p<0.001), the most compelling effect was observed for genes related to the cell cycle (e.g., Cdc20, Ccnb) (Fig. 3A). In addition, glucagon receptor (Gcgr) expression was also decreased (p=0.0004) (Fig. 3B). Combining these data with a proteinprotein interaction network model, we identified two functional networks that were upregulated relevant to the current study (Fig. 4, circles): one associated with hormone secretion (e.g. Ppy, Pyy), and the other with extracellular structure [e.g. Sdc1 (that encodes for syndecan 1), Hs3st3b1 (that encodes for heparan sulfate-glucosamine 3-sulfotransferase 3B1)] (Fig. 3B and Fig. 4). The amplification of pancreatic heparan sulfate that was observed in hep-tg mice (Fig. 3C) could be attributed, at least in part, to the increase in Sdc1 and Hs3st3b1. Among the downregulated genes, a highly connected network of genes related to the cell cycle function was recognized (Fig. 4, box). Altogether, these data suggest that its ability to

Page 12 of 85 modulate gene expression is consistent with the role of heparanase as a potent regulator of islet structure and function. Heparanase Transgenic Mice Exhibit Resistance To Multiple Low-Dose Streptozotocin- Induced Hyperglycemia As hep-tg islets are enriched in heparan sulfate, which is important for beta cell survival (13), we attempted to chemically induce diabetes in these mice. Interestingly, although WT animals showed robust hyperglycemia within one week of STZ injection, hep-tg mice failed to present with plasma glucose levels comparable to WT (Fig. 5A). Measurement of plasma insulin provided one explanation for this observation. STZ caused a precipitous drop of plasma insulin in WT animals, an effect that was absent in hep-tg mice (Fig. 5B). Prolonging the duration of the study did not change the results - hep-tg mice remained resistant to STZinduced hyperglycemia for up to 8 weeks following injection (Supplementary Fig. 6). This resistance to STZ was likely not due to any difference in expression of pancreatic GLUT2 (Supplementary Fig. 7), the transporter required for STZ uptake. To establish whether the failure to develop STZ-induced hyperglycemia was a consequence of beta cell preservation, we quantified beta cell mass. Surprisingly, upon injection of STZ, both the WT and hep-tg mice displayed an identical loss of insulin-positive cells (Fig. 5C). This implies that the resistance to experimental diabetes in hep-tg mice is not a consequence of beta cell survival, consistent with observations seen in GRKO mice, which do not develop Type 1 diabetes when injected with STZ (20). Given our observation that hep-tg mice secrete more insulin in response to an OGTT (Fig. 1F), it is possible that the islets, which survived STZ toxicity in hep-tg diabetic mice, have a greater insulin secretory capacity. In this regard, hep-tg mice injected with STZ were still, albeit in limited capacity, able to secrete insulin in response to an IPGTT (Fig. 5D). Taken together, our data suggest that the protective effects of heparanase against STZ-induced diabetes is unlikely to be a consequence of HS-mediated beta cell

Page 13 of 85 survival, but could be related to the ability of any residual beta cells in these animals to continue their secretion of insulin. Severity Of Remains Disparate Between WT and Hep-tg Mice Injected With Single High Dose Streptozotocin Multiple low-dose STZ induces beta cell death by activating immune mechanisms (22). Hence, we also tested the response of hep-tg mice to a single high dose of STZ, which produces severe diabetes by direct beta cell toxicity via DNA alkylation (23). In contrast to our observation with multiple low-dose STZ, both WT and hep-tg mice injected with 200 mg/kg STZ (single high dose) exhibited a significant reduction in plasma insulin levels (Fig. 6B). In agreement with the loss of insulin, WT diabetic mice demonstrated sustained hyperglycemia (Fig. 6A). Significantly, the values from day 3 onwards for these mice were beyond the upper limit of detection (33.3 mmol/l) of the glucometer, suggesting that the actual blood glucose concentrations were likely higher. Although high dose STZ was competent to lower insulin and induce diabetes in hep-tg mice, the magnitude of hyperglycemia was lower than that seen in WT animals (Fig. 6A), suggesting the contribution of an insulin mimetic factor. Fibroblast growth factor-21 (FGF21) has significant beneficial effects on glucose homeostasis (34). Measurement of FGF21 plasma concentration indicated that hep-tg mice had higher concentrations compared to WT (Fig. 6C). Moreover, although STZ-induced diabetes caused a significant drop in FGF21 levels in WT mice, its levels in hep-tg mice was strikingly amplified by three-fold (Fig. 6C), and was likely related to its increased hepatic production (Supplementary Fig. 8). Another hormone that has glucose-lowering effects independent of insulin is glucagon-like peptide 1 (GLP-1) (35). In response to high dose STZ, GLP-1 trended in the opposite direction between these two strains (Fig. 6D). Thus, in hep-tg mice, driving circulating insulin concentrations down to very low levels uncovers novel protective pathways for the management of hyperglycemia.

Page 14 of 85 Discussion Homozygous transgenic mice overexpressing human heparanase globally are fertile and have a normal life span (10). In addition, physiological functions associated with heparanase in these mice include roles in embryonic implantation, food consumption, tissue remodeling, and vascularization (10). In the current study, an unexpected feature of these animals was our discovery of improved glucose homeostasis, but resistance to glucagon. Strikingly, compared to controls, hep-tg mice manifested significant changes in islet gene expression of over 2000 genes. These mice also demonstrated resistance to chemically-induced diabetes. Our data suggest a novel role for heparanase in mechanisms that serve to correct hyperglycemia. In hep-tg mice, both basal insulin and glucose levels were lower than in WT following a 6 h fast, suggesting superior insulin sensitivity in these animals. This idea was reinforced by an enhanced insulin response to activation of skeletal muscle Akt in this study. Supporting these observations is that, in myeloma cells, insulin receptor is the predominant receptor tyrosine kinase activated by heparanase (6). The lower glucose concentration was evident in the presence of high glucagon levels, suggesting a deficit in glucagon action. Interestingly, an insulin tolerance test used to evaluate insulin efficiency was unable to detect any difference in blood glucose lowering between the two groups. Moreover, the traditional oral glucose challenge test was also unable to confirm a higher rate of glucose disposal in hep-tg mice. We reasoned that the overnight fast prior to performing the OGTT could explain this inconsistency. Following fasting, hypersensitivity of the liver to glucagon accounts for a multi-fold increase in hepatic glucose production (35). Should this happen, even marginally, in hep-tg mice that have higher basal glucagon levels, the advantage of augmented insulin sensitivity in these animals would be dampened, resulting in a similar glucose clearance compared to WT. Remarkably, in hep-tg mice, the OGTT elicited higher insulin release, whereas isolated islets from these animals had a more robust secretion of insulin in response to glucose, even with

Page 15 of 85 islet insulin content and gene expression remaining unchanged. Our data suggest that although heparanase appears to enhance insulin signalling, whole body tests to determine insulin sensitivity were unable to confirm this, likely as a consequence of the overriding influence of an insulin counter-regulatory hormone such as glucagon in the hep-tg mice. In GRKO mice, a prolonged deficiency in glucagon receptor signalling activates alpha cell proliferation, and leads to a compensatory elevation in glucagon production (36). We were surprised to observe that these characteristics in GRKO mice were strikingly similar to features seen in hep-tg animals, which did not respond to a glucagon challenge as well as WT mice. More importantly, hep-tg islets presented an architecture that is conducive to hypersecretion of glucagon, under basal conditions and also subsequent to injection of arginine. In these mice, alpha cells lost their peripheral mantle-like localization and were distributed randomly throughout the islets. In addition, the alpha/beta cell ratio was higher in hep-tg mice. These changes in alpha cells were evident even though islet size and size distribution remained unaffected. Currently, it is unclear whether these alterations in alpha cell morphology are consequences of glucagon resistance, or of disrupted pancreatic islet development through the action of heparanase on HS. Measurement of HS in the pancreas produced an unanticipated result; rather than depleting HS, the hep-tg pancreas displayed the opposing phenotype, with increased expression of HS, and the gene (Hs3st3b1) that encodes for its biosynthetic enzyme, 3-O-sulfotransferase, being dominant features. A similar observation has also been made in the hep-tg liver, where the authors described the accumulation of heparin-like HS degradation products, both within and between cells, resulting in an accelerated biosynthesis of HS (37). It follows that like in the liver, pancreatic heparanase processes HS into smaller heparin-like oligosaccharides. Regardless of whether a result of its enzymatic activities, or its non-traditional signalling activities, overexpression of heparanase produced a dramatic alteration in the islet

cells using immunostaining indicated that hep-tg beta cells were destroyed to an equal extent Page 16 of 85 transcriptome. Among the clusters of genes that were increased, of particular interest were genes that regulate cell death, synthesize extracellular matrix components, and produce pancreatic hormones. Related to cell death, this was not unusual as cancer cells use the properties of heparanase to induce gene expression and cell survival (38). The impact on the extracellular matrix is especially meaningful, as components like collagen and laminin have roles in islet cell differentiation, proliferation, and hormone secretion (39). Finally, peptide hormones like somatostatin and pancreatic polypeptide hormone are known to influence pancreatic islet function in a paracrine manner (40; 41). Unlike the upregulated genes that were enriched for diverse functions, the downregulated genes formed a coherent functional group including those tightly linked to the cell cycle. Whether these changes in expression creates an energy-sparing environment that would be conducive to a higher level of hormone production, as recently reported (42), is certainly consistent with our observations and deserving of further investigation. Collectively, our data provide support for the notion that heparanase in the pancreas is particularly beneficial, reinforcing it against the effects of exogenous stress. Multiple low-dose STZ mimics Type 1 diabetes by stimulating beta cell apoptosis through the recruitment of immune cells (22). Given the role that HS plays in modulating the innate immune response by effecting immune cell adhesion, cytokine and chemokine binding, and providing a physical barrier against leukocytes infiltration (43), an increase in HS as seen in hep-tg mice could be potentially beneficial. This, when added to the genomic signature in these mice that is protective against cell death, one would anticipate a resistance to MLD-STZ beta cell cytotoxicity. Indeed, our results, for the first time, demonstrate that hep-tg animals developed less severe hyperglycemia compared to WT, and that this was associated with an almost unchanged circulating insulin concentration. Inexplicably, the preserved plasma insulin in these animals was not a consequence of higher beta cell mass, as quantification of these

Page 17 of 85 compared to WT. It is possible that, of the beta cells that survived STZ toxicity in both groups, an enhanced insulin secretory capacity in hep-tg animals could explain this anomaly. In support, hep-tg diabetic animals can still increase insulin secretion in response to a glucose challenge, an effect that is lost in WT diabetic mice. Our data suggest that heparanase produces beta cells that are more functionally efficient, an effect likely related to an elevation of 3-O-sulfotransferases and accumulation of HS in these islets. STZ in high doses induces diabetes by a mechanism different from MLD-STZ; it directly destroys beta cells by alkylating DNA (23). Additionally, as there is a comparatively more severe and rapid development of hyperglycemia related to a catastrophic loss of insulin, we tested whether heparanase would continue to correct hyperglycemia in this model. Despite the identical low levels of residual circulating insulin in both diabetic groups, hep-tg mice continued to have significantly lower blood glucose. In GRKO mice, FGF21 and GLP-1 have been suggested to be the hormones responsible for glucose clearance in the absence of insulin (44). This was also true in the hep-tg diabetic mouse. In these animals, regardless of their basal concentrations, SHD-STZ caused an augmentation of both FGF21 and GLP-1, effects that can contribute to their lower glucose levels. Altogether, the results indicate that, with the onset of hyperglycemia, additional glucose lowering mechanisms are triggered in hep-tg mice. It has been proposed that HS in pancreatic islet BM functions as an obstruction against leukocyte infiltration, in addition to protecting the beta cell against ROS induced cell death (45). Thus, in the non-obese diabetic (NOD) mouse, augmented production of active heparanase by immune cells permits destruction of islet BM HS, entry of leukocytes, and degradation of intracellular HS. As the heparanase inhibitor PI-88 preserved intra-islet HS and protected NOD mice from T1D, the authors concluded that heparanase inhibition is useful against T1D progression (13). Contrasting results were observed in NOD mice injected with

Page 18 of 85 exogenous heparanase, which ameliorated the occurrence of diabetes (46). Unlike the NOD mice or metastatic cancer cells (47) where the overproduction of active heparanase is responsible for disease development, our model is one in which, in a disease free background, latent heparanase is overexpressed, with an associated amplification of islet HS. Thus, although active heparanase has been considered a pathogenic marker, our study has discovered multiple novel properties of latent heparanase related to the control of glycaemia. Overexpression of this protein caused glucose lowering, potentiation of insulin secretion, HS induction, prodigious changes in islet gene expression, and protection against chemically induced diabetes. This study unlocks the possibility of utilizing these properties of heparanase in the management of diabetes.

Page 19 of 85 Article Information Supplementary Material. This article contains Supplementary Data online at http://diabetes.diabetesjournals.org. Funding. This work was supported by an operating grant from the Canadian Institutes of Health Research to BR (CIHR-MOP-133547). Amy Pei-Ling Chiu and Dahai Zhang are the recipients of Doctoral Student Research Awards from the Canadian Association. Duality of Interest. None declared. Author Contributions. D.Z. performed most of the experiments and with B.R., generated the hypothesis, designed the study, performed the data analysis and wrote the manuscript. F.W., N.L., A.P.C., A.W., J.J., D.B., F.T., A.A., B.H., and B.H. contributed in part, to some of the experiments performed. S.F., S.S., and C.N did RNA Sequencing and its analysis. T.P., I.V., J.D.J., and T.J.K. contributed to the discussion and manuscript editing. B.R. is the guarantor of this work.

Page 20 of 85 Figure Legends Fig. 1. Heparanase overexpression improves glucose homeostasis. A. PCR amplification of DNA extracted from WT and hep-tg mice ear tissue. L19 is used as an internal control. B-C. Following a 6 h fast, plasma insulin and blood glucose levels were measured in WT and TG mice. n=9-10, p<0.05 vs. WT. D. Intraperitoneal administration of insulin (2 µg/kg body weight) to 6 h fasted mice, followed by skeletal muscle isolation for Western Blot. n=3, p<0.05 vs. control, # p<0.05 vs. WT insulin. E-F. Following an overnight fast (16 h), WT and TG mice were administered an oral glucose (2 g/kg) gavage. At the indicated times, blood glucose and insulin were measured. n=4-5, p<0.05 vs. WT. G. Isolated islets (10) from the different groups were used to extract insulin with Acid-Ethanol, and the insulin content was measured with ELISA (WT, 2.38±0.23 ng/ml; hep-tg, 3.08±1.04 ng/ml, p=0.218). Different islets (10) from the same animals were exposed to 3 mm and 16.7 mm glucose respectively, and insulin secretion determined. n=3, p<0.05. Fig. 2. Glucagon resistance and altered islet cytoarchitecture in heparanase overexpressing mice. A. Plasma glucagon levels in the two groups of mice were determined after a 16 h fast. n=4-5, p<0.05 vs. WT. B. For the glucagon challenge, 16 h fasted mice were administered glucagon (1 µg/kg), and blood glucose measured at the indicated times. n=5, p<0.05 vs. WT. C. In 6 h fasted mice, 2 g/kg L-arginine was administered i.p. and plasma glucagon, insulin, and blood glucose (fold change in glucose was calculated by normalizing to the corresponding value at t=0) measured at the indicated times. n=5, p<0.05 vs. WT. D. Higher magnification of insulin and glucagon positive cells in pancreas isolated from the two groups of animals using immunofluorescence staining, together with quantification of the alpha/beta cell ratio, n=3, p<0.05 vs. WT. E. Whole pancreas sections were stained for insulin (red) and glucagon (green), scanned, and islets counted and quantified by size.

Page 21 of 85 Figure 3. Islet transcriptome changes dramatically in hep-tg mice. A. Islet RNA from WT and hep-tg mice was sequenced and the (p<0.001) upregulated (red) and downregulated (green) genes were categorized based on the functions. Bar represents the enrichment of the genes in particular category over the total genes of input. B. Representative genes in extracellular matrix, signaling and cell cycle were shown. Heparanase gene was used as positive control of the entire RNA sequencing, together with islet protein detection as inset. C. Pancreas staining for heparan sulfate using alcian blue (ph 5.8, 0.65 M MgCl 2 ) and its quantification using Western blot. n=3, p<0.05 vs. WT. Fig. 4. Association network of genes that were significantly different between WT and hep-tg mice. Analysis of a protein-protein interaction network assembled from RNAseq data. The blue square depicts the entire downregulated network that is related to cell cycle. The red circles encompass both the hormone-associated network and the extracellular elements network. Lines represent associations based on differential expression evidence. Fig. 5. Hep-tg mice are resistant to multiple low-dose streptozotocin induced diabetes. A-B. To induce Type 1 diabetes, WT and hep-tg mice were administered 50 mg/kg STZ for 5 consecutive days. 7 days after the last STZ injection and following a 6 h fast, tail vein blood was used for glucose and plasma insulin determination, n=5-10, p<0.05. C. Pancreas from STZ injected mice was isolated and immune-stained for insulin (red) and glucagon (green), and insulin positive cells were quantified in the different groups. n=3, p<0.05. vs con. D. Following an IPGTT, plasma insulin in STZ injected WT and hep-tg mice were measured over the indicated times. The data is expressed as a fold change normalized to the corresponding values at t=0 min for each animal within the two groups. n=3-5, p<0.05 vs WT. Fig. 6. Single high dose streptozotocin in hep-tg mice uncovers protective responses to manage hyperglycemia. A. A single high dose (200 mg/kg) STZ (SHD-STZ) was administered i.p., and glucose levels monitored daily over 5 days. n=4-8, p<0.05. B-D. One

Page 22 of 85 week after injection of SHD-STZ, insulin, FGF21 and GLP-1 levels from WT and hep-tg mice were determined. n=4-8, p<0.05. References 1. Sarrazin S, Lamanna WC, Esko JD: Heparan sulfate proteoglycans. Cold Spring Harb Perspect Biol 2011;3 2. Gittes GK: Developmental biology of the pancreas: a comprehensive review. Dev Biol 2009;326:4-35 3. Purushothaman A, Hurst DR, Pisano C, Mizumoto S, Sugahara K, Sanderson RD: Heparanase-mediated loss of nuclear syndecan-1 enhances histone acetyltransferase (HAT) activity to promote expression of genes that drive an aggressive tumor phenotype. J Biol Chem 2011;286:30377-30383 4. Wu L, Viola CM, Brzozowski AM, Davies GJ: Structural characterization of human heparanase reveals insights into substrate recognition. Nat Struct Mol Biol 2015;22:1016-1022 5. Vlodavsky I, Ilan N, Naggi A, Casu B: Heparanase: Structure, biological functions, and inhibition by heparin-derived mimetics of heparan sulfate. Curr Pharm Des 2007;13:2057-2073 6. Purushothaman A, Babitz SK, Sanderson RD: Heparanase Enhances the Insulin Receptor Signaling Pathway to Activate Extracellular Signal-regulated Kinase in Multiple Myeloma. J Biol Chem 2012;287:41288-41296 7. Riaz A, Ilan N, Vlodavsky I, Li JP, Johansson S: Characterization of Heparanase-induced Phosphatidylinositol 3-Kinase-AKT Activation and Its Integrin Dependence. J Biol Chem 2013;288:12366-12375 8. Zetser A, Bashenko Y, Edovitsky E, Levy-Adam F, Vlodavsky I, Ilan N: Heparanase induces vascular endothelial growth factor expression: Correlation with p38 phosphorylation levels and Src activation. Cancer Res 2006;66:1455-1463 9. Zetser A, Bashenko Y, Edovitsky E, Levy-Adam F, Vlodavsky I, Ilan N: Heparanase induces vascular endothelial growth factor expression: correlation with p38 phosphorylation levels and Src activation. Cancer Res 2006;66:1455-1463 10. Zcharia E, Metzger S, Chajek-Shaul T, Aingorn H, Elkin M, Friedmann Y, Weinstein T, Li JP, Lindahl U, Vlodavsky I: Transgenic expression of mammalian heparanase uncovers physiological functions of heparan sulfate in tissue morphogenesis, vascularization, and feeding behavior. FASEB J 2004;18:252-263 11. Arvatz G, Weissmann M, Ilan N, Vlodavsky I: Heparanase and cancer progression: New directions, new promises. Hum Vaccin Immunother 2016:1-4 12. Bogdani M, Korpos E, Simeonovic CJ, Parish CR, Sorokin L, Wight TN: Extracellular matrix components in the pathogenesis of type 1 diabetes. Curr Diab Rep 2014;14:552 13. Ziolkowski AF, Popp SK, Freeman C, Parish CR, Simeonovic CJ: Heparan sulfate and heparanase play key roles in mouse beta cell survival and autoimmune diabetes. J Clin Invest 2012;122:132-141 14. Wan A, Rodrigues B: Endothelial Cell-Cardiomyocyte Crosstalk in Diabetic Cardiomyopathy. Cardiovasc Res 2016; 15. Assady S, Alter J, Axelman E, Zohar Y, Sabo E, Litvak M, Kaplan M, Ilan N, Vlodavsky I, Abassi Z: Nephroprotective effect of heparanase in experimental nephrotic syndrome. PLoS One 2015;10:e0119610 16. Jendresen CB, Cui H, Zhang X, Vlodavsky I, Nilsson LN, Li JP: Overexpression of heparanase lowers the amyloid burden in amyloid-beta precursor protein transgenic mice. J Biol Chem 2015;290:5053-5064 17. Purushothaman A, Chen LG, Yang Y, Sanderson RD: Heparanase Stimulation of Protease Expression Implicates It as a Master Regulator of the Aggressive Tumor Phenotype in Myeloma. J Biol Chem 2008;283:32628-32636 18. Aston-Mourney K, Zraika S, Udayasankar J, Subramanian SL, Green PS, Kahn SE, Hull RL: Matrix metalloproteinase-9 reduces islet amyloid formation by degrading islet amyloid polypeptide. J Biol Chem 2013;288:3553-3559 19. Meier DT, Tu LH, Zraika S, Hogan MF, Templin AT, Hull RL, Raleigh DP, Kahn SE: Matrix Metalloproteinase-9 Protects Islets from Amyloid-induced Toxicity. J Biol Chem 2015;290:30475-30485 20. Sorensen H, Winzell MS, Brand CL, Fosgerau K, Gelling RW, Nishimura E, Ahren B: Glucagon receptor knockout mice display increased insulin sensitivity and impaired beta-cell function. 2006;55:3463-3469

21. Conarello SL, Jiang G, Mu J, Li Z, Woods J, Zycband E, Ronan J, Liu F, Roy RS, Zhu L, Charron MJ, Zhang BB: Glucagon receptor knockout mice are resistant to diet-induced obesity and streptozotocin-mediated beta cell loss and hyperglycaemia. Diabetologia 2007;50:142-150 22. McEvoy RC, Andersson J, Sandler S, Hellerstrom C: Multiple low-dose streptozotocin-induced diabetes in the mouse. Evidence for stimulation of a cytotoxic cellular immune response against an insulinproducing beta cell line. J Clin Invest 1984;74:715-722 23. Deeds MC, Anderson JM, Armstrong AS, Gastineau DA, Hiddinga HJ, Jahangir A, Eberhardt NL, Kudva YC: Single dose streptozotocin-induced diabetes: considerations for study design in islet transplantation models. Lab Anim 2011;45:131-140 24. Asadi A, Bruin JE, Kieffer TJ: Characterization of Antibodies to Products of Proinsulin Processing Using Immunofluorescence Staining of Pancreas in Multiple Species. J Histochem Cytochem 2015;63:646-662 25. An D, Pulinilkunnil T, Qi D, Ghosh S, Abrahani A, Rodrigues B: The metabolic "switch" AMPK regulates cardiac heparin-releasable lipoprotein lipase. Am J Physiol Endocrinol Metab 2005;288:E246-253 26. Alejandro EU, Kalynyak TB, Taghizadeh F, Gwiazda KS, Rawstron EK, Jacob KJ, Johnson JD: Acute Insulin Signaling in Pancreatic Beta-Cells Is Mediated by Multiple Raf-1 Dependent Pathways. Endocrinology 2010;151:502-512 27. Love MI, Huber W, Anders S: Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15:550 28. Wang L, Wang S, Li W: RSeQC: quality control of RNA-seq experiments. Bioinformatics 2012;28:2184-2185 29. Grabherr MG, Haas BJ, Yassour M, Levin JZ, Thompson DA, Amit I, Adiconis X, Fan L, Raychowdhury R, Zeng QD, Chen ZH, Mauceli E, Hacohen N, Gnirke A, Rhind N, di Palma F, Birren BW, Nusbaum C, Lindblad- Toh K, Friedman N, Regev A: Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat Biotechnol 2011;29:644-U130 30. Zimmer Y, Milo-Landesman D, Svetlanov A, Efrat S: Genes induced by growth arrest in a pancreatic beta cell line: identification by analysis of cdna arrays. FEBS Lett 1999;457:65-70 31. Oropeza D, Jouvet N, Bouyakdan K, Perron G, Ringuette LJ, Philipson LH, Kiss RS, Poitout V, Alquier T, Estall JL: PGC-1 coactivators in beta-cells regulate lipid metabolism and are essential for insulin secretion coupled to fatty acids. Mol Metab 2015;4:811-822 32. Sabatini PV, Krentz NAJ, Zarrouki B, Westwell-Roper CY, Nian CL, Uy RA, Shapiro AMJ, Poitout V, Lynn FC: Npas4 Is a Novel Activity-Regulated Cytoprotective Factor in Pancreatic beta-cells. 2013;62:2808-2820 33. Lingohr MK, Buettner R, Rhodes CJ: Pancreatic beta-cell growth and survival - a role in obesity-linked type 2 diabetes? Trends Mol Med 2002;8:375-384 34. Kwon MM, O'Dwyer SM, Baker RK, Covey SD, Kieffer TJ: FGF21-Mediated Improvements in Glucose Clearance Require Uncoupling Protein 1. Cell Rep 2015;13:1521-1527 35. Dardevet D, Moore MC, Neal D, DiCostanzo CA, Snead W, Cherrington AD: Insulin-independent effects of GLP-1 on canine liver glucose metabolism: duration of infusion and involvement of hepatoportal region. Am J Physiol Endocrinol Metab 2004;287:E75-81 36. Gelling RW, Du XQ, Dichmann DS, Romer J, Huang H, Cui L, Obici S, Tang B, Holst JJ, Fledelius C, Johansen PB, Rossetti L, Jelicks LA, Serup P, Nishimura E, Charron MJ: Lower blood glucose, hyperglucagonemia, and pancreatic alpha cell hyperplasia in glucagon receptor knockout mice. Proc Natl Acad Sci U S A 2003;100:1438-1443 37. Escobar Galvis ML, Jia J, Zhang X, Jastrebova N, Spillmann D, Gottfridsson E, van Kuppevelt TH, Zcharia E, Vlodavsky I, Lindahl U, Li JP: Transgenic or tumor-induced expression of heparanase upregulates sulfation of heparan sulfate. Nat Chem Biol 2007;3:773-778 38. Nadir Y, Brenner B: Heparanase procoagulant activity in cancer progression. Thromb Res 2016;140 Suppl 1:S44-48 39. Meda P: Protein-mediated interactions of pancreatic islet cells. Scientifica (Cairo) 2013;2013:621249 40. Hauge-Evans AC, King AJ, Carmignac D, Richardson CC, Robinson ICAF, Low MJ, Christie MR, Persaud SJ, Jones PM: Somatostatin Secreted by Islet delta-cells Fulfills Multiple Roles as a Paracrine Regulator of Islet Function. 2009;58:403-411 41. Kahleova H, Mari A, Nofrate V, Matoulek M, Kazdova L, Hill M, Pelikanova T: Improvement in beta-cell function after diet-induced weight loss is associated with decrease in pancreatic polypeptide in subjects with type 2 diabetes. J Complications 2012;26:442-449 Page 23 of 85

Page 24 of 85 42. Szabat M, Page MM, Panzhinskiy E, Skovso S, Mojibian M, Fernandez-Tajes J, Bruin JE, Bround MJ, Lee JTC, Xu EE, Taghizadeh F, O'Dwyer S, van de Bunt M, Moon KM, Sinha S, Han J, Fan Y, Lynn FC, Trucco M, Borchers CH, Foster LJ, Nislow C, Kieffer TJ, Johnson JD: Reduced Insulin Production Relieves Endoplasmic Reticulum Stress and Induces beta Cell Proliferation. Cell Metabolism 2016;23:179-193 43. Simon Davis DA, Parish CR: Heparan sulfate: a ubiquitous glycosaminoglycan with multiple roles in immunity. Front Immunol 2013;4:470 44. Omar BA, Andersen B, Hald J, Raun K, Nishimura E, Ahren B: Fibroblast growth factor 21 (FGF21) and glucagon-like peptide 1 contribute to diabetes resistance in glucagon receptor-deficient mice. 2014;63:101-110 45. Simeonovic CJ, Ziolkowski AF, Wu Z, Choong FJ, Freeman C, Parish CR: Heparanase and autoimmune diabetes. Front Immunol 2013;4:471 46. Bitan M, Weiss L, Zeira M, Reich S, Pappo O, Vlodavsky I, Slavin S: Heparanase prevents the development of type 1 diabetes in non-obese diabetic mice by regulating T-cell activation and cytokines production. -Metab Res 2008;24:413-421 47. Vlodavsky I, Friedmann Y: Molecular properties and involvement of heparanase in cancer metastasis and angiogenesis. J Clin Invest 2001;108:341-347

Page 25 of 85 A L19 Hep WT TG WT TG TG L19+Hep L19 Hep Plasma Insulin B (ng/ml) 1.2 1.0 0.8 0.6 0.4 0.2 0.0 WT TG Blood Glucose C (mmol/l) 12 10 8 6 4 2 0 WT TG D Skeletal Muscle E Insulin pakt Ser473 _ WT + _ TG + 20 total AKT 16 beta-actin pakt/akt (fold increase) 14 - Insulin 12 + Insulin 10 8 6 4 2 0 # OGTT Glucose (mmol/l) 12 8 4 0 WT TG 0 30 60 90 120 (min) WT TG F G OGTT Insulin (ng/ml) 2.0 1.6 1.2 0.8 WT TG Insulin (ng/ml) 2.5 2.0 1.5 1.0 3 mm 16.7mM 0.4 0.5 0.0 0 30 60 90 120 (min) 0.0 WT TG Fig. 1

Page 26 of 85 A Plasma glucagon (pg/ml) 400 300 200 100 0 WT TG B Blood Glucose (mmol/l) 8.0 6.0 4.0 Glucagon WT TG C Glucagon (pg/ml) 2500 2000 1500 1000 500 Arginine Insulin (ng/ml) 0.55 0.50 0.45 0.40 Arginine WT TG 0 20 40 60 80 100 120 Glucose (fold change) 1.2 0.9 Arginine 0.6 (min) D 0 0 15 60 90 Glucagon Insulin (min) 0.35 0 15 60 90 (min) 0.20 Alpha/beta Cells Ratio 0.15 0.10 0.05 0 15 60 90 (min) WT TG 50 m 0.00 WT TG E Glucagon Insulin WT Average Islet Size ( m 2 ) 8000 6000 4000 2000 0 0.6 WT TG Frequency (%) 0.5 0.4 0.3 0.2 0.1 WT TG 0.0 <300 300-5000 >5000 ( m 2 ) TG 250 m Islet Sizes 40 m Fig. 2

Page 27 of 85 A Cellular process Development Cell Death Homeostasis Cell cycle Cellular Structure single-organism process single-organism cellular process tissue development regulation of cell death regulation of apoptotic process regulation of programmed cell death digestion positive regulation of biological process organ development regulation of biological quality response to stress cell communication chemical homeostasis biological regulation response to stimulus small molecule metabolic process positive regulation of cellular process organic hydroxy compound metabolic process homeostatic process organic acid metabolic process spindle organization spindle assembly regulation of microtubule cytoskeleton organization regulation of chromosome segregation regulation of attachment of spindle microtubules to kinetochore regulation of cell cycle process regulation of mitotic cell cycle mitotic nuclear division microtubule cytoskeleton organization chromosome segregation microtubule cytoskeleton organization involved in mitosis sister chromatid segregation nuclear chromosome segregation organelle fission mitotic cell cycle process single-organism organelle organization cell cycle process nuclear division mitotic sister chromatid segregation cell cycle cell division 1176 985 upregulated downregulated -60-40 -20 0 20 40 60 80 Enrichment (% of input) B Log2 fold change C 6 4 2 0-2 Hpse Acacb Myo5c 5 m 75 kda 50 kda Gal Hspb1 Igf1 Col7a1 WT Gcg Ppy Lama2 WT TG Lamb3 Ngfr Pyy Sdc1 Rgs4 Tubb1 Tuba1c Latent Heparanase Active Heparanase TG HS Beta- Actin Apoe Hs3st3b1 Npas4 WT Gcgr TG Signaling Structural molecule Nek2 Ccna2 Ccnb1 Nusap1 Cdk1 Pbk Aspm Ckap2l Cdc20 Top2a HS/beta-actin (fold change) 4 3 2 1 0 WT Enzymes Cell Cycle TG Espl1 Cenpf Fig. 3

Page 28 of 85 Fig. 4

Page 29 of 85 Con MLD-STZ A Plasma glucose (mmol/l) 25 20 15 10 5 B Plasma Insulin (ng/ml) 1.2 0.8 0.4 C 0 WT TG WT TG 0.0 400 M WT-MLD-STZ TG-MLD-STZ 0.006 WT-MLD-STZ TG-MLD-STZ Insulin positive/total area 0.005 0.004 0.003 0.002 0.001 0.000 D 2.0 1.8 WT-STZ TG-STZ WT TG Insulin (fold change) 1.6 1.4 1.2 1.0 0.8 0.6 0.4 0 min 15 min 90 min Fig. 5