Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lucy Marzban is active.

Publication


Featured researches published by Lucy Marzban.


Experimental Gerontology | 2003

Islet amyloid polypeptide and type 2 diabetes

Lucy Marzban; Kirily Park; C. Bruce Verchere

Type 2 diabetes is associated with progressive beta-cell failure manifest as a decline in insulin secretion and increasing hyperglycemia. A growing body of evidence suggests that beta-cell failure in type 2 diabetes correlates with the formation of pancreatic islet amyloid deposits, indicating that islet amyloid may have an important role in beta-cell loss in this disease. Islet amyloid polypeptide (IAPP; amylin), the major component of islet amyloid, is co-secreted with insulin from beta-cells. In type 2 diabetes, this peptide aggregates to form amyloid fibrils that are toxic to beta-cells. The mechanism(s) responsible for islet amyloid formation in type 2 diabetes is still unclear but it appears that an increase in the secretion of IAPP, per se, is not sufficient. Other factors, such as impairment in the processing of proIAPP, the IAPP precursor, have been proposed to contribute to the development of islet amyloid deposits. Inhibitors of islet amyloid fibril formation might prevent the progression to beta-cell failure in type 2 diabetes and should therefore be considered as a therapeutic approach to treat this disease.


Diabetes | 2006

Impaired NH2-Terminal Processing of Human Proislet Amyloid Polypeptide by the Prohormone Convertase PC2 Leads to Amyloid Formation and Cell Death

Lucy Marzban; Christopher J. Rhodes; Donald F. Steiner; Leena Haataja; Philippe A. Halban; C. Bruce Verchere

Islet amyloid, formed by aggregation of islet amyloid polypeptide (IAPP; amylin), is a pathological characteristic of the pancreas in type 2 diabetes and may contribute to the progressive loss of β-cells in this disease. We tested the hypothesis that impaired processing of the IAPP precursor proIAPP contributes to amyloid formation and cell death. GH3 cells lacking the prohormone convertase 1/3 (PC1/3) and IAPP and with very low levels of prohormone convertase 2 (PC2) were transduced with adenovirus (Ad) expressing human or rat (control) proIAPP linked to green fluorescent protein, with or without Ad-PC2 or Ad-PC1/3. Expression of human proIAPP increased the number of transferase-mediated dUTP nick-end labeling (TUNEL)-positive cells 96 h after transduction (+hIAPP 8.7 ± 0.4% vs. control 3.0 ± 0.4%; P < 0.05). COOH-terminal processing of human proIAPP by PC1/3 increased (hIAPP+PC1/3 10.4 ± 0.7%; P < 0.05), whereas NH2-terminal processing of proIAPP by addition of PC2 markedly decreased (hIAPP+PC2 5.5 ± 0.5%; P < 0.05) the number of apoptotic GH3 cells. Islets from mice lacking PC2 and with β-cell expression of human proIAPP (hIAPP+/+/PC2−/−) developed amyloid associated with β-cell death during 2-week culture. Rescue of PC2 expression by ex vivo transduction with Ad-PC2 restored NH2-terminal processing to mature IAPP and decreased both the extent of amyloid formation and the number of TUNEL-positive cells (−PC2 26.5 ± 4.1% vs. +PC2 16.1 ± 4.3%; P < 0.05). These findings suggest that impaired NH2-terminal processing of proIAPP leads to amyloid formation and cell death and that accumulation of the NH2-terminally extended human proIAPP intermediate may be a critical initiating step in amyloid formation.


Diabetes | 2008

Small Interfering RNA–Mediated Suppression of Proislet Amyloid Polypeptide Expression Inhibits Islet Amyloid Formation and Enhances Survival of Human Islets in Culture

Lucy Marzban; Alejandra Tomas; Thomas C. Becker; Lawrence Rosenberg; José Oberholzer; Paul E. Fraser; Philippe A. Halban; C. Bruce Verchere

OBJECTIVE—Islet amyloid, formed by aggregation of the β-cell peptide islet amyloid polypeptide (IAPP; amylin), is a pathological characteristic of pancreatic islets in type 2 diabetes. Toxic IAPP aggregates likely contribute to the progressive loss of β-cells in this disease. We used cultured human islets as an ex vivo model of amyloid formation to investigate whether suppression of proIAPP expression would inhibit islet amyloid formation and enhance β-cell survival and function. RESEARCH DESIGN AND METHODS—Islets from cadaveric organ donors were transduced with a recombinant adenovirus expressing a short interfering RNA (siRNA) designed to suppress human proIAPP (Ad-hProIAPP-siRNA), cultured for 10 days, and then assessed for the presence of islet amyloid, β-cell apoptosis, and β-cell function. RESULTS—Thioflavine S–positive amyloid deposits were clearly present after 10 days of culture. Transduction with Ad-hProIAPP-siRNA reduced proIAPP expression by 75% compared with nontransduced islets as assessed by Western blot analysis of islet lysates 4 days after transduction. siRNA-mediated inhibition of IAPP expression decreased islet amyloid area by 63% compared with nontransduced cultured islets. Cell death assessed by transferase-mediated dUTP nick-end labeling staining was decreased by 50% in transduced cultured human islets, associated with a significant increase in islet insulin content (control, 100 ± 4 vs. +Ad-siRNA, 153 ± 22%, P < 0.01) and glucose-stimulated insulin secretion (control, 222 ± 33 vs. +Ad-siRNA, 285 ± 21 percent basal, P < 0.05). CONCLUSIONS—These findings demonstrate that inhibition of IAPP synthesis prevents amyloid formation and β-cell death in cultured human islets. Inhibitors of IAPP synthesis may have therapeutic value in type 2 diabetes.


Diabetologia | 2012

Deletion of Fas protects islet beta cells from cytotoxic effects of human islet amyloid polypeptide

Y. J. Park; S. Lee; Timothy J. Kieffer; Garth L. Warnock; Nooshin Safikhan; M. Speck; Z. Hao; M. Woo; Lucy Marzban

Aims/hypothesisIslet amyloid, which is mainly composed of human islet amyloid polypeptide (hIAPP), is a pathological characteristic of type 2 diabetes and also forms in cultured and transplanted islets. We used islet beta cells as well as two ex vivo models of islet amyloid formation, cultured human islets and hIAPP-expressing transgenic mouse islets with or without beta cell Fas deletion, to test whether: (1) the aggregation of endogenous hIAPP induces Fas upregulation in beta cells; and (2) deletion or blocking of Fas protects beta cells from amyloid toxicity.MethodsINS-1, mouse or human islet cells were cultured with hIAPP alone, or with amyloid inhibitor or Fas antagonist. Non-transduced islets, and human islets or hIAPP-expressing mouse islets transduced with an adenovirus that delivers a human proIAPP-specific small interfering RNA (siRNA) (Ad-ProhIAPP-siRNA) were cultured to form amyloid. Mouse islets expressing hIAPP with or without Fas were similarly cultured. Beta cell Fas upregulation, caspase-3 activation, apoptosis and function, and islet IL-1β levels were assessed.ResultshIAPP treatment induced Fas upregulation, caspase-3 activation and apoptosis in INS-1 and islet cells. The amyloid inhibitor or Fas antagonist reduced apoptosis in hIAPP-treated beta cells. Islet cells with Fas deletion had lower hIAPP-induced beta cell apoptosis than those expressing Fas. Ad-ProhIAPP-siRNA-mediated amyloid inhibition reduced Fas upregulation and IL-1β immunoreactivity in human and hIAPP-expressing mouse islets. Cultured hIAPP-expressing mouse islets with Fas deletion had similar amyloid levels, but lower caspase-3 activation and beta cell apoptosis, and a higher islet beta:alpha cell ratio and insulin response to glucose, compared with islets expressing Fas and hIAPP.Conclusions/interpretationThe aggregation of biosynthetic hIAPP produced in islets induces beta cell apoptosis, at least partially, via Fas upregulation and the Fas-mediated apoptotic pathway. Deletion of Fas protects islet beta cells from the cytotoxic effects of endogenously secreted (and exogenously applied) hIAPP.


Diabetologia | 2010

Differences between amyloid toxicity in alpha and beta cells in human and mouse islets and the role of caspase-3

E. Law; S. Lu; Timothy J. Kieffer; Garth L. Warnock; Ziliang Ao; M. Woo; Lucy Marzban

Aims/hypothesisType 2 diabetes is characterised by decreased beta cell mass and islet amyloid formation. Islet amyloid formed by aggregation of human islet amyloid polypeptide (hIAPP) is associated with beta cell apoptosis. We used human and transgenic mouse islets in culture to examine whether deletion of caspase-3 protects islets from apoptosis induced by endogenously produced and exogenously applied hIAPP and compared hIAPP toxicity in islet alpha and beta cells.MethodsHuman and wild-type or caspase-3 knockout mouse islet cells were treated with hIAPP. Rat insulinoma INS-1 cells were similarly cultured with hIAPP and the amyloid inhibitor Congo Red or caspase-3 inhibitor. Human and hIAPP-expressing caspase-3 knockout mouse islets were cultured to form amyloid fibrils and assessed for beta and alpha cell apoptosis, beta cell function and caspase-3 activation.ResultshIAPP-treated INS-1 cells had increased caspase-3 activation and apoptosis, both of which were reduced by inhibitors of amyloid or caspase-3. Similarly, hIAPP-treated human and mouse islet beta cells had elevated active caspase-3- and TUNEL-positive cells, whereas mouse islet cells lacking caspase-3 had markedly lower beta cell but comparable alpha cell apoptosis. During culture, human islets that formed amyloid had higher active caspase-3- and TUNEL-positive beta cells than those without detectable amyloid. Finally, cultured hIAPP-expressing mouse islets lacking caspase-3 had markedly lower beta cell apoptosis than those expressing caspase-3, associated with an increase in islet beta cell/alpha cell ratio, insulin content and glucose response.Conclusions/interpretationPrevention of caspase-3 activation protects islet beta cells from apoptosis induced by fibrillogenesis of endogenously secreted and exogenously applied hIAPP. Islet beta cells are more susceptible to hIAPP toxicity than alpha cells cultured under the same conditions.


Diabetes | 2009

Cleavage of Protein Kinase D After Acute Hypoinsulinemia Prevents Excessive Lipoprotein Lipase–Mediated Cardiac Triglyceride Accumulation

Min Suk Kim; Fang Wang; Prasanth Puthanveetil; Girish Kewalramani; Sheila M. Innis; Lucy Marzban; Susan F. Steinberg; Travis D. Webber; Timothy J. Kieffer; Brian Rodrigues

OBJECTIVE During hypoinsulinemia, when cardiac glucose utilization is impaired, the heart rapidly adapts to using more fatty acids. One means by which this is achieved is through lipoprotein lipase (LPL). We determined the mechanisms by which the heart regulates LPL after acute hypoinsulinemia. RESEARCH DESIGN AND METHODS We used two different doses of streptozocin (55 [d-55] and 100 [d-100] mg/kg) to induce moderate and severe hypoinsulinemia, respectively, in rats. Isolated cardiomyocytes were also used for transfection or silencing of protein kinase D (PKD) and caspase-3. RESULTS There was substantial increase in LPL in d-55 hearts, an effect that was absent in severely hypoinsulinemic d-100 animals. Measurement of PKD, a key element involved in increasing LPL, revealed that only d-100 hearts showed an increase in proteolysis of PKD, an effect that required activation of caspase-3 together with loss of 14-3-3ζ, a binding protein that protects enzymes against degradation. In vitro, phosphomimetic PKD colocalized with LPL in the trans-golgi. PKD, when mutated to prevent its cleavage by caspase-3 and silencing of caspase-3, was able to increase LPL activity. Using a caspase inhibitor (Z-DEVD) in d-100 animals, we effectively lowered caspase-3 activity, prevented PKD cleavage, and increased LPL vesicle formation and translocation to the vascular lumen. This increase in cardiac luminal LPL was associated with a striking accumulation of cardiac triglyceride in Z-DEVD–treated d-100 rats. CONCLUSIONS After severe hypoinsulinemia, activation of caspase-3 can restrict LPL translocation to the vascular lumen. When caspase-3 is inhibited, this compensatory response is lost, leading to lipid accumulation in the heart.


American Journal of Pathology | 2012

Three-Dimensional Scaffolds Reduce Islet Amyloid Formation and Enhance Survival and Function of Cultured Human Islets

Yun Zhang; Reza B. Jalili; Garth L. Warnock; Ziliang Ao; Lucy Marzban; Aziz Ghahary

Islet transplantation provides a promising approach for treatment of type 1 diabetes mellitus. Amyloid formation and loss of extracellular matrix are two nonimmune factors contributing to death of isolated human islets. We tested the effects of two types of three-dimensional scaffolds, collagen matrix (CM) and fibroblast-populated collagen matrix (FPCM), on amyloid formation, viability, and function of isolated islets. Islets from cadaveric donors were cultured in FPCM, CM, or two-dimensional plate (2D) for 7 days. After 7 days, compared with the 2D culture condition, CM and FPCM markedly reduced amyloid formation of cultured islets and decreased apoptotic β-cell rate by ∼75%. IL-1β and Fas levels were also reduced in scaffold-embedded islets. Furthermore, β/α cell ratios were increased by ∼18% and ∼36% in CM- and FPCM-embedded islets, respectively. Insulin content and insulin response to elevated glucose were also enhanced by both three-dimensional scaffolds. Moreover, culture in CM and FPCM (but not 2D) preserved insulin, GLUT-2, and PDX-1 mRNA expression. FPCM-embedded islets had significantly higher insulin response and lower amyloid formation than CM-embedded islets. These findings suggest that three-dimensional scaffolds reduce amyloid formation and improve viability and function of human islets in vitro, and that CM and fibroblasts have additive effects in enhancing islet function and reducing amyloid formation. Using this strategy is likely to improve outcome in human islet transplantation.


Molecular and Cellular Biochemistry | 2001

In vivo effects of insulin and bis(maltolato)oxovanadium (IV) on PKB activity in the skeletal muscle and liver of diabetic rats

Lucy Marzban; Sanjay Bhanot; John H. McNeill

In this study, the in vivo effects of insulin and chronic treatment with bis(maltolato)oxovanadium (IV) (BMOV) on protein kinase B (PKB) activity were examined in the liver and skeletal muscle from two animal models of diabetes, the STZ‐diabetic Wistar rat and the fatty Zucker rat. Animals were treated with BMOV in the drinking water (0.75–1 mg/ml) for 3 (or 8) weeks and sacrificed with or without insulin injection. Insulin (5 U/kg, i.v.) increased PKBα activity more than 10‐fold and PKBβ activity more than 3‐fold in both animal models. Despite the development of insulin resistance, insulin‐induced activation of PKBα was not impaired in the STZ‐diabetic rats up to 9 weeks of diabetes, excluding a role for PKBα in the development of insulin resistance in type 1 diabetes. Insulin-induced PKBα activity was markedly reduced in the skeletal muscle of fatty Zucker rats as compared to lean littermates (fatty: 7‐fold vs. lean: 14‐fold). In contrast, a significant increase in insulin‐stimulated PKBa activity was observed in the liver of fatty Zucker rats (fatty: 15.7‐fold vs. lean: 7.6‐fold). Chronic treatment with BMOV normalized plasma glucose levels in STZ‐diabetic rats and decreased plasma insulin levels in fatty Zucker rats but did not have any effect on basal or insulin‐induced PKBα and PKBβ activities. In conclusion (i) in STZ‐diabetic rats PKB activity was normal up to 9 weeks of diabetes; (ii) in fatty Zucker rats insulin‐induced activation of PKBα (but not PKBβ) was markedly altered in both tissues; (iii) changes in PKBα activity were tissue specific; (iv) the glucoregulatory effects of BMOV were independent of PKB activity.


Diabetologia | 2014

The role of caspase-8 in amyloid-induced beta cell death in human and mouse islets

Yoo Jin Park; Minna Woo; Timothy J. Kieffer; Razqallah Hakem; Nooshin Safikhan; Fan Yang; Ziliang Ao; Garth L. Warnock; Lucy Marzban

Aims/hypothesisReduced beta cell mass due to increased beta cell apoptosis is a key defect in type 2 diabetes. Islet amyloid, formed by the aggregation of human islet amyloid polypeptide (hIAPP), contributes to beta cell death in type 2 diabetes and in islet grafts in patients with type 1 diabetes. In this study, we used human islets and hIAPP-expressing mouse islets with beta cell Casp8 deletion to (1) investigate the role of caspase-8 in amyloid-induced beta cell apoptosis and (2) test whether caspase-8 inhibition protects beta cells from amyloid toxicity.MethodsHuman islet cells were cultured with hIAPP alone, or with caspase-8, Fas or amyloid inhibitors. Human islets and wild-type or hIAPP-expressing mouse islets with or without caspase-8 expression (generated using a Cre/loxP system) were cultured to form amyloid. Caspase-8 and -3 activation, Fas and FLICE inhibitory protein (FLIP) expression, islet beta cell and amyloid area, IL-1β levels, and the beta:alpha cell ratio were assessed.ResultshIAPP treatment induced activation of caspase-8 and -3 in islet beta cells (via Fas upregulation), resulting in apoptosis, which was markedly reduced by blocking caspase-8, Fas or amyloid. Amyloid formation in cultured human and hIAPP-expressing mouse islets induced caspase-8 activation, which was associated with Fas upregulation and elevated islet IL-1β levels. hIAPP-expressing mouse islets with Casp8 deletion had comparable amyloid, IL-1β and Fas levels with those expressing hIAPP and Casp8, but markedly lower beta cell apoptosis, higher beta:alpha cell ratio, greater beta cell area, and enhanced beta cell function.Conclusions/interpretationBeta cell Fas upregulation by endogenously produced and exogenously applied hIAPP aggregates promotes caspase-8 activation, resulting in beta cell apoptosis. The prevention of amyloid-induced caspase-8 activation enhances beta cell survival and function in islets.


Diabetes, Obesity and Metabolism | 2017

Dual role of interleukin-1β in islet amyloid formation and its β-cell toxicity: Implications for type 2 diabetes and islet transplantation

Yoo Jin Park; Garth L. Warnock; Ziliang Ao; Nooshin Safikhan; Mark Meloche; Ali Asadi; Timothy J. Kieffer; Lucy Marzban

Islet amyloid, formed by aggregation of human islet amyloid polypeptide (hIAPP), contributes to β‐cell failure in type 2 diabetes, cultured and transplanted islets. We previously showed that biosynthetic hIAPP aggregates induce β‐cell Fas upregulation and activation of the Fas apoptotic pathway. We used cultured human and hIAPP‐expressing mouse islets to investigate: (1) the role of interleukin‐1β (IL‐1β) in amyloid‐induced Fas upregulation; and (2) the effects of IL‐1β‐induced β‐cell dysfunction on pro‐islet amyloid polypeptide (proIAPP) processing and amyloid formation.

Collaboration


Dive into the Lucy Marzban's collaboration.

Top Co-Authors

Avatar

Garth L. Warnock

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Ziliang Ao

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Timothy J. Kieffer

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Yoo Jin Park

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

C. Bruce Verchere

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Mark Meloche

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Nooshin Safikhan

Vancouver General Hospital

View shared research outputs
Top Co-Authors

Avatar

Yun Zhang

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aziz Ghahary

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge