Network


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

Hotspot


Dive into the research topics where Travis D. Webber is active.

Publication


Featured researches published by Travis D. Webber.


Gastroenterology | 2010

Glucose-Dependent Insulinotropic Polypeptide Is Expressed in Pancreatic Islet α-Cells and Promotes Insulin Secretion

Yukihiro Fujita; Rhonda D. Wideman; Ali Asadi; Gary K. Yang; Robert K. Baker; Travis D. Webber; Tianjiao Zhang; Rennian Wang; Ziliang Ao; Garth L. Warnock; Yin Nam Kwok; Timothy J. Kieffer

BACKGROUND & AIMS Glucose-dependent insulinotropic polypeptide (GIP) and the proglucagon product glucagon-like peptide-1 (GLP-1) are gastrointestinal hormones that are released in response to nutrient intake and promote insulin secretion. Interestingly, a subset of enteroendocrine cells express both GIP and GLP-1. We sought to determine whether GIP also might be co-expressed with proglucagon in pancreatic alpha-cells. METHODS We assessed GIP expression via reverse-transcription polymerase chain reaction, in situ hybridization, and immunohistochemistry. We developed a novel bioassay to measure GIP release from isolated islets, compared the biological activities of full-length and truncated GIP, and assessed the impact of immunoneutralization of islet GIP on glucose-stimulated insulin secretion in isolated islets. RESULTS GIP messenger RNA was present in mouse islets; GIP protein localized to islet alpha-cells of mouse, human, and snake pancreas, based on immunohistochemical analyses. However, using a C-terminal GIP antibody, immunoreactivity was detected in islets from prohormone convertase (PC) 2 knockout but not wild-type mice. Bioactive GIP was secreted from mouse and human islets after arginine stimulation. In the perfused mouse pancreas, GIP(1-42) and amidated GIP(1-30) had equipotent insulinotropic actions. Finally, immunoneutralization of GIP secreted by isolated islets decreased glucose-stimulated insulin secretion. CONCLUSIONS GIP is expressed in and secreted from pancreatic islets; in alpha-cells, PC2 processes proGIP to yield a truncated but bioactive form of GIP that differs from the PC1/3-derived form from K-cells. Islet-derived GIP promotes islet glucose competence and also could support islet development and/or survival.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Improving function and survival of pancreatic islets by endogenous production of glucagon-like peptide 1 (GLP-1)

Rhonda D. Wideman; Irene L. Y. Yu; Travis D. Webber; C. Bruce Verchere; James D. Johnson; Anthony T. Cheung; Timothy J. Kieffer

Glucagon-like peptide 1 (GLP-1) is a hormone that has received significant attention as a therapy for diabetes because of its ability to stimulate insulin biosynthesis and release and to promote growth and survival of insulin-producing β cells. While GLP-1 is produced from the proglucagon precursor by means of prohormone convertase (PC) 1/3 activity in enteroendocrine L cells, the same precursor is differentially processed by PC2 in pancreatic islet α cells to release glucagon, leaving GLP-1 trapped within a larger fragment with no known function. We hypothesized that we could induce GLP-1 production directly within pancreatic islets by means of delivery of PC1/3 and, further, that this intervention would improve the viability and function of islets. Here, we show that adenovirus-mediated expression of PC1/3 in α cells increases islet GLP-1 secretion, resulting in improved glucose-stimulated insulin secretion and enhanced survival in response to cytokine treatment. PC1/3 expression in α cells also improved performance after islet transplantation in a mouse model of type 1 diabetes, possibly by enhancing nuclear Pdx1 and insulin content of islet β cells. These results demonstrate a unique strategy for liberating GLP-1 from directly within the target organ and highlight the potential for up-regulating islet GLP-1 production as a means of treating diabetes.


PLOS ONE | 2013

Initial cell seeding density influences pancreatic endocrine development during in vitro differentiation of human embryonic stem cells.

Blair K. Gage; Travis D. Webber; Timothy J. Kieffer

Human embryonic stem cells (hESCs) have the ability to form cells derived from all three germ layers, and as such have received significant attention as a possible source for insulin-secreting pancreatic beta-cells for diabetes treatment. While considerable advances have been made in generating hESC-derived insulin-producing cells, to date in vitro-derived glucose-responsive beta-cells have remained an elusive goal. With the objective of increasing the in vitro formation of pancreatic endocrine cells, we examined the effect of varying initial cell seeding density from 1.3 x 104 cells/cm2 to 5.3 x 104 cells/cm2 followed by a 21-day pancreatic endocrine differentiation protocol. Low density-seeded cells were found to be biased toward the G2/M phases of the cell cycle and failed to efficiently differentiate into SOX17-CXCR4 co-positive definitive endoderm cells leaving increased numbers of OCT4 positive cells in day 4 cultures. Moderate density cultures effectively formed definitive endoderm and progressed to express PDX1 in approximately 20% of the culture. High density cultures contained approximately double the numbers of PDX1 positive pancreatic progenitor cells and also showed increased expression of MNX1, PTF1a, NGN3, ARX, and PAX4 compared to cultures seeded at moderate density. The cultures seeded at high density displayed increased formation of polyhormonal pancreatic endocrine cell populations co-expressing insulin, glucagon and somatostatin. The maturation process giving rise to these endocrine cell populations followed the expected cascade of pancreatic progenitor marker (PDX1 and MNX1) expression, followed by pancreatic endocrine specification marker expression (BRN4, PAX4, ARX, NEUROD1, NKX6.1 and NKX2.2) and then pancreatic hormone expression (insulin, glucagon and somatostatin). Taken together these data suggest that initial cell seeding density plays an important role in both germ layer specification and pancreatic progenitor commitment, which precedes pancreatic endocrine cell formation. This work highlights the need to examine standard culture variables such as seeding density when optimizing hESC differentiation protocols.


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.


Journal of Molecular Medicine | 2009

Treatment of diabetes by transplantation of drug-inducible insulin-producing gut cells

Suraj Unniappan; Rhonda D. Wideman; Christine Donald; Virginia Gunn; Jennifer L. Wall; Qiu-Xia Zhang; Travis D. Webber; Anthony T. Cheung; Timothy J. Kieffer

Most patients with type 1 diabetes rely on multiple daily insulin injections to maintain blood glucose control. However, insulin injections carry the risk of inducing hypoglycemia and do not eliminate diabetic complications. We sought to develop and evaluate a regulatable cell-based system for delivery of insulin to treat diabetes. We generated two intestinal cell lines in which human insulin expression is controlled by mifepristone. Insulin mRNA expression was dependent on the mifepristone dose and incubation time and cells displayed insulin and C-peptide immunoreactivity and glucose-induced insulin release following mifepristone treatment. Cell transplantation followed by mifepristone administration reversed streptozotocin (STZ)-induced diabetes in mice, and this effect was dependent on the mifepristone dose delivered. These data support the notion that engineering regulatable insulin expression within a cell already equipped for regulated secretion may be efficacious for the treatment of insulin-dependent diabetes.


Differentiation | 2010

Cellular reprogramming of human amniotic fluid cells to express insulin.

Blair K. Gage; Michael J. Riedel; Francis Karanu; Alireza Rezania; Yukihiro Fujita; Travis D. Webber; Robert K. Baker; Rhonda D. Wideman; Timothy J. Kieffer

Islet transplantation represents a potential cure for type 1 diabetes; however, a lack of sufficient donor material limits its clinical use. To address the shortfall of islet availability, surrogate insulin-producing cells are sought. Studies suggest that human amniotic fluid (hAF) contains multipotent progenitor cells capable of differentiating to all three germ layers. Here, we used high-content, live-cell imaging to assess the ability to reprogram hAF cells towards a beta cell phenotype. A fluorescent reporter system was developed where DsRed express (DSRE) expression is driven by the human insulin promoter. Using integrative lentiviral technology, we created stable reporter hAF cells that could be routinely monitored for insulin promoter activation. These cells were subjected to combinatorial high-content screening using adenoviral-mediated expression of up to six transcription factors important for beta cell development. Cells were monitored for DSRE expression which revealed an optimal combination of the transcription factors required to induce insulin gene expression in hAF cells. These optimally induced cells were examined for expression of additional beta cell transcription factors and proteins involved in glucose sensing and insulin processing. RT-qPCR revealed very low level expression of insulin that was ultimately insufficient to reverse streptozotocin-induced diabetes following sub-capsular kidney transplantation. High-content, live-cell imaging using fluorescent reporter cells provides a convenient method for repeated assessment of cellular reprogramming. hAF cells could be reprogrammed to express key beta cell proteins, however insulin gene expression was insufficient to reverse hyperglycemia in diabetic animals.


Molecular Therapy | 2008

Treatment of Obesity and Diabetes in Mice by Transplant of Gut Cells Engineered to Produce Leptin

Sarah N Oosman; Ada W Lam; George Harb; Suraj Unniappan; Ni T. Lam; Travis D. Webber; Daniel Bruch; Qiu-Xia Zhang; Gregory S. Korbutt; Timothy J. Kieffer

Leptin injections evoke weight loss by causing a reduction in food consumption and an increase in energy expenditure. Also, the administration of leptin lowers blood glucose levels in some rodent models of diabetes and in humans with lipodystrophy. We explored the therapeutic potential of delivering leptin to obese, diabetic ob/ob mice and to mice fed on a high-fat diet (HFD), by transplanting gut-derived cells engineered to produce leptin, under the regulation of an inducing agent, mifepristone. These cells expressed and released leptin in a mifepristone dose-dependent and time-dependent manner. The engineered cells were either transplanted into the mice under the kidney capsule or were encapsulated in alginate and injected into the intraperitoneal cavity, while mifepristone was delivered by implanting 14-day release pellets. In ob/ob mice, leptin delivery by this method caused a significant reduction in food intake and profound weight loss, which was controllable by adjusting the dose of mifepristone. These transplants also achieved rapid and persistent amelioration of diabetes. However, mice fed on a HFD were resistant to the leptin therapy. These results indicate that gut cells can be modified to express leptin in an inducible manner and that the transplantation of these cells has a therapeutic effect in leptin-deficient mice, but not in mice fed on a HFD.


PLOS ONE | 2015

The Role of ARX in Human Pancreatic Endocrine Specification.

Blair K. Gage; Ali Asadi; Robert K. Baker; Travis D. Webber; Rennian Wang; Masayuki Itoh; Masaharu Hayashi; Rie Miyata; Takumi Akashi; Timothy J. Kieffer

The in vitro differentiation of human embryonic stem cells (hESCs) offers a model system to explore human development. Humans with mutations in the transcription factor Aristaless Related Homeobox (ARX) often suffer from the syndrome X-linked lissencephaly with ambiguous genitalia (XLAG), affecting many cell types including those of the pancreas. Indeed, XLAG pancreatic islets lack glucagon and pancreatic polypeptide-positive cells but retain somatostatin, insulin, and ghrelin-positive cells. To further examine the role of ARX in human pancreatic endocrine development, we utilized genomic editing in hESCs to generate deletions in ARX. ARX knockout hESCs retained pancreatic differentiation capacity and ARX knockout endocrine cells were biased toward somatostatin-positive cells (94% of endocrine cells) with reduced pancreatic polypeptide (rarely detected), glucagon (90% reduced) and insulin-positive (65% reduced) lineages. ARX knockout somatostatin-positive cells shared expression patterns with human fetal and adult δ-cells. Differentiated ARX knockout cells upregulated PAX4, NKX2.2, ISL1, HHEX, PCSK1, PCSK2 expression while downregulating PAX6 and IRX2. Re-expression of ARX in ARX knockout pancreatic progenitors reduced HHEX and increased PAX6 and insulin expression following differentiation. Taken together these data suggest that ARX plays a key role in pancreatic endocrine fate specification of pancreatic polypeptide, somatostatin, glucagon and insulin positive cells from hESCs.


American Journal of Physiology-endocrinology and Metabolism | 2009

Incretin release from gut is acutely enhanced by sugar but not by sweeteners in vivo

Yukihiro Fujita; Rhonda D. Wideman; Madeleine Speck; Ali Asadi; David S. King; Travis D. Webber; Masakazu Haneda; Timothy J. Kieffer


Cell Metabolism | 2006

The pancreatic β cell is a key site for mediating the effects of leptin on glucose homeostasis

Scott D. Covey; Rhonda D. Wideman; Christine McDonald; Suraj Unniappan; Frank K. Huynh; Ali Asadi; Madeleine Speck; Travis D. Webber; Streamson C. Chua; Timothy J. Kieffer

Collaboration


Dive into the Travis D. Webber's collaboration.

Top Co-Authors

Avatar

Timothy J. Kieffer

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Rhonda D. Wideman

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Ali Asadi

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Blair K. Gage

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Robert K. Baker

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Suraj Unniappan

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Yukihiro Fujita

Asahikawa Medical University

View shared research outputs
Top Co-Authors

Avatar

Madeleine Speck

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge