Network


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

Hotspot


Dive into the research topics where M. Lamb is active.

Publication


Featured researches published by M. Lamb.


Cell Transplantation | 2014

In Vitro Maturation of Viable Islets From Partially Digested Young Pig Pancreas

M. Lamb; Laugenour K; Ouwen Liang; Michael Alexander; Clarence E. Foster; Jonathan R. T. Lakey

Isolation of islets from market-sized pigs is costly, with considerable islet losses from fragmentation occurring during isolation and tissue culture. Fetal and neonatal pigs yield insulin unresponsive islet-like cell clusters that become glucose-responsive after extended periods of time. Both issues impact clinical applicability and commercial scale-up. We have focused our efforts on a cost-effective scalable method of isolating viable insulin-responsive islets. Young Yorkshire pigs (mean age 20 days, range 4–30 days) underwent rapid pancreatectomy (<5 min) and partial digestion using low-dose collagenase, followed by in vitro culture at 37°C and 5% CO2 for up to 14 days. Islet viability was assessed using FDA/PI or Newport Green, and function was assessed using a glucose-stimulated insulin release (GSIR) assay. Islet yield was performed using enumeration of dithizonestained aliquots. The young porcine (YP) islet yield at dissociation was 12.6 ± 2.1 × 103 IEQ (mean ± SEM) per organ and increased to 33.3 ± 6.4 × 103 IEQ after 7 days of in vitro culture. Viability was 97.3 ± 7% at dissociation and remained over 90% viable after 11 days in tissue culture (n = ns). Glucose responsiveness increased throughout maturation in culture. The stimulation index (SI) of the islets increased from 1.7 ± 2 on culture day 3 to 2.58 ± 0.5 on culture day 7. These results suggest that this method is both efficient and scalable for isolating and maturing insulin-responsive porcine islets in culture.


Biomaterials | 2014

Noninvasive evaluation of the vascular response to transplantation of alginate encapsulated islets using the dorsal skin-fold model

Rahul Krishnan; Rajan P. Arora; Michael Alexander; Sean M. White; M. Lamb; Clarence E. Foster; Bernard Choi; Jonathan R. T. Lakey

Alginate encapsulation reduces the risk of transplant rejection by evading immune-mediated cell injury and rejection; however, poor vascular perfusion results in graft failure. Since existing imaging models are incapable of quantifying the vascular response to biomaterial implants after transplantation, in this study, we demonstrate the use of in vivo laser speckle imaging (LSI) and wide-field functional imaging (WiFI) to monitor the microvascular environment surrounding biomaterial implants. The vascular response to two islet-containing biomaterial encapsulation devices, alginate microcapsules and a high-guluronate alginate sheet, was studied and compared after implantation into the mouse dorsal window chamber (N = 4 per implant group). Images obtained over a 14-day period using LSI and WiFI were analyzed using algorithms to quantify blood flow, hemoglobin oxygen saturation and vascular density. Using our method, we were able to monitor the changes in the peri-implant microvasculature noninvasively without the use of fluorescent dyes. Significant changes in blood flow, hemoglobin oxygen saturation and vascular density were noted as early as the first week post-transplant. The dorsal window chamber model enables comparison of host responses to transplanted biomaterials. Future experiments will study the effect of changes in alginate composition on the vascular and immune responses.


Transplantation proceedings | 2011

Function and Viability of Human Islets Encapsulated in Alginate Sheets: In Vitro and in Vivo Culture

M. Lamb; Richard Wood Storrs; S. Li; O. Liang; Laugenour K; Randy Dorian; D. Chapman; Hirohito Ichii; David K. Imagawa; Clarence E. Foster; Scott R. King; Jonathan R. T. Lakey

Islet encapsulation offers an immune system barrier for islet transplantation, and encapsulation within an alginate sheetlike structure offers the ability to be retrievable after transplanted. This study aims to show that human islets encapsulated into islet sheets remain functional and viable after 8 weeks in culture or when transplanted into the subcutaneous space of rats. Human islets were isolated from cadaveric organs. Dissociation and purification were done using enzymatic digestion and a continuous Ficoll-UWD gradient. Purified human islets were encapsulated in alginate sheets. Human Islet sheets were either kept in culture, at 37°C and 5% CO(2), or transplanted subcutaneously into Lewis rats. After 1, 2, 4, and 8 weeks, the human islet sheets were retrieved from the rats and assessed. The viability of the sheets was measured using fluorescein diacetate (FDA)/propidium iodide (PI), and function was measured through glucose-stimulated insulin release, in which the sheets were incubated for an hour in low-glucose concentration (2.8 mmol/L) and then high (28 mmol/L), then high (28 mmol/L) plus 3-isobutyl-1-methylxanthine (50 μm). Human islet sheets remained both viable, above 70%, and functional, with a stimulation index (insulin secretion in high glucose divided by insulin secretion in low glucose) above 1.5, over 8 weeks of culture or subcutaneous transplantation. Islet transplantation continues to make advances in the treatment of type 1 diabetes. These preliminary results suggest that encapsulated islets sheets can survive and maintain islet viability and function in vivo, within the subcutaneous region.


Islets | 2013

Young porcine endocrine pancreatic islets cultured in fibrin show improved resistance toward hydrogen peroxide.

Carina Kuehn; Jonathan R. T. Lakey; M. Lamb; Patrick Vermette

Aim: To study the protective effect of a fibrin scaffold toward embedded young porcine endocrine pancreatic islets from hydrogen peroxide within the context of islet encapsulation in transplantation. Methods: After isolation and in vitro maturation, groups of 200 young porcine islet equivalents (IEQ) were embedded in a 200 µL fibrin gel and exposed to 2 concentrations (10 and 100 µM) of hydrogen peroxide (H2O2) to investigate the ability of fibrin to protect islets against apoptotic stimuli. As a control, young porcine islets were seeded in tissue culture polystyrene (TCPS) well plates and exposed to the same H2O2 concentrations. Islet integrity, viability and function were then investigated. Results: Morphologically, the integrity of islets embedded in fibrin gels was better preserved compared with that of islets cultured in TCPS plates, when exposed to H2O2. Immunofluorescence staining showed that insulin and glucagon expression was higher in islets cultured in fibrin. Overall, H2O2 incubation led to decreased insulin and glucagon expression. A TUNEL assay revealed elevated numbers of apoptotic cells for islets cultured in TCPS plates when compared with those embedded in fibrin. Islets cultured in TCPS plates and exposed to H2O2 had diminished ability to secrete insulin in response to glucose stimulation, whereas islets embedded in fibrin maintained their glucose responsiveness. Insulin trapped in fibrin was extracted and quantified, revealing insulin in the extract. Conclusions/Interpretation: Fibrin has a protective effect on young porcine endocrine pancreatic islets exposed to hydrogen peroxide.


Islets | 2012

GPR54 peptide agonists stimulate insulin secretion from murine, porcine and human islets

James E. Bowe; Victoria L. Foot; Stephanie A. Amiel; Guo Cai Huang; M. Lamb; Jonathan R. T. Lakey; Peter M. Jones; Shanta J. Persaud

This study was designed to determine the effects of 10 and 13 amino acid forms of kisspeptin on dynamic insulin secretion from mammalian islets since it is not clear from published data whether the shorter peptide is stimulatory while the longer peptide inhibits insulin release. Insulin secretion was measured by radioimmunoassay following perifusion of human, pig, rat and mouse isolated islets with kisspeptin-10 or kisspeptin-13 in the presence of 20 mM glucose. Both peptides stimulated rapid, reversible potentiation of glucose-stimulated insulin secretion from islets of all species tested. These data indicate that both kisspeptin-10 and kisspeptin-13, which is an extension of kisspeptin-10 by three amino acids, act directly at islet β-cells of various species to potentiate insulin secretion, and suggest that inhibitory effects reported in earlier studies may reflect differences in experimental protocols.


Cell Transplantation | 2014

Current status of islet encapsulation.

Lourdes Robles; Storrs R; M. Lamb; Michael Alexander; Lakey

Cell encapsulation is a method of encasing cells in a semipermeable matrix that provides a permeable gradient for the passage of oxygen and nutrients, but effectively blocks immune-regulating cells from reaching the graft, preventing rejection. This concept has been described as early as the 1930s, but it has exhibited substantial achievements over the last decade. Several advances in encapsulation engineering, chemical purification, applications, and cell viability promise to make this a revolutionary technology. Several obstacles still need to be overcome before this process becomes a reality, including developing a reliable source of islets or insulin-producing cells, determining the ideal biomaterial to promote graft function, reducing the host response to the encapsulation device, and ultimately a streamlined, scaled-up process for industry to be able to efficiently and safely produce encapsulated cells for clinical use. This article provides a comprehensive review of cell encapsulation of islets for the treatment of type 1 diabetes, including a historical perspective, current research findings, and future studies.


Transplantation Proceedings | 2014

Impact of Hypothermic Preservation on Tissue Yield and Viability in Pig Pancreata

Michael Alexander; Rahul Krishnan; B. Buder; M. Lamb; Laugenour K; D. Chapman; Clarence E. Foster; Jonathan R. T. Lakey


Cellular and Molecular Bioengineering | 2014

Culturing Free-Floating and Fibrin-Embedded Islets with Endothelial Cells: Effects on Insulin Secretion and Apoptosis

Evan A. Dubiel; Jonathan R. T. Lakey; M. Lamb; Patrick Vermette


Archive | 2015

Islet Xenotransplantation: An Update on Recent Advances and Future Prospects 45

Rahul Krishnan; M. Lamb; Michael Alexander; David K. Imagawa; Jonathan R. T. Lakey


Transplantation | 2012

A Comparison of the Host Response to Two Types of Biomaterial Encapsulated Islets Using the Dorsal Window Chamber Model: 2370

R. Krishnan; R. Arora; M. Lamb; O. Liang; S. M. White; R. Storrs; R. Dorian; S. King; B. Choi; Jonathan R. T. Lakey

Collaboration


Dive into the M. Lamb's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laugenour K

University of California

View shared research outputs
Top Co-Authors

Avatar

Rahul Krishnan

University of California

View shared research outputs
Top Co-Authors

Avatar

D. Chapman

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge