Network


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

Hotspot


Dive into the research topics where Vijayaganapathy Vaithilingam is active.

Publication


Featured researches published by Vijayaganapathy Vaithilingam.


Current Opinion in Organ Transplantation | 2008

Islet cell transplantation

Vijayaganapathy Vaithilingam; Gayathri Sundaram; Bernard E. Tuch

Purpose of reviewThe transplantation of human islets has come a long way since the first diabetic person became insulin independent in 1989. The advent of a steroid-free immunosuppressive protocol in 2000 resulted in most recipients becoming insulin independent and remaining so for a year. However, β-cell function declines thereafter. Strategies to enhance the islet mass transplanted and preserve β-cell function are necessary. Recent findingsThis review covers recent advances in determining the selection of appropriate enzymes for islet isolation, use of pancreases from heart-dead donors and techniques for predicting the functional capacity of isolated islets prior to transplantation. Changing the transplantation site away from the liver, where many islets are destroyed by an inflammatory process, is reviewed, and the possibility of seeding islets onto three-dimensional biodegradable scaffolds discussed. A method of preventing apoptosis of the β cells prior to transplantation is detailed, as is the beneficial effect of using exenatide, after transplantation. Novel techniques to image islets are discussed, and this requires the labelling of the islets prior to implantation. Enhancing the vascularization of islets is shown to enhance functional outcomes. Encapsulation of the islets should obviate the need for using antirejection drugs, and it may be possible to expand β cells in vitro. SummaryThe above strategies are likely to enhance the outcomes of clinical islet transplants.


Biomaterials | 2011

Effect of alginate encapsulation on the cellular transcriptome of human islets

Vijayaganapathy Vaithilingam; Nayeem Quayum; Mugdha V. Joglekar; Jan Jensen; Anandwardhan A. Hardikar; Jose Oberholzer; Gilles J. Guillemin; Bernard E. Tuch

Encapsulation of human islets may prevent their immune rejection when transplanted into diabetic recipients. To assist in understanding why clinical outcomes with encapsulated islets were not ideal, we examined the effect of encapsulation on their global gene (mRNA) and selected miRNAs (non-coding (nc)RNA) expression. For functional studies, encapsulated islets were transplanted into peritoneal cavity of diabetic NOD-SCID mice. Genomics analysis and transplantation studies demonstrate that islet origin and isolation centres are a major source of variation in islet quality. In contrast, tissue culture and the encapsulation process had only a minimal effect, and did not affect islet viability. Microarray analysis showed that as few as 29 genes were up-regulated and 2 genes down-regulated (cut-off threshold 0.1) by encapsulation. Ingenuity analysis showed that up-regulated genes were involved mostly in inflammation, especially chemotaxis, and vascularisation. However, protein expression of these factors was not altered by encapsulation, raising doubts about the biosignificance of the gene changes. Encapsulation had no effect on levels of islet miRNAs. In vivo studies indicate differences among the centres in the quality of the islets isolated. We conclude that microencapsulation of human islets with barium alginate has little effect on their transcriptome.


Pancreas | 2011

Functional capacity of human islets after long-distance shipment and encapsulation.

Vijayaganapathy Vaithilingam; Barbara Barbaro; Jose Oberholzer; Bernard E. Tuch

Objective: Human islets produced at an isolation center are shipped to researchers, usually taking short periods to arrive at their destination. In this study, we investigated whether human islets after long-distance shipment across the Pacific Ocean for 2 to 3 days and encapsulation could maintain their functionality. Methods: Human islets were encapsulated in alginate and viability assessed using carboxyfluorescein diacetate and propidium iodide. Stimulation index after static glucose incubation was calculated. Streptozotocin-induced diabetic nonobese diabetic/severe combined immunodeficient mice were transplanted with 3000, 2000, or 1000 islet equivalents of nonencapsulated and encapsulated islets and glucose levels monitored. When levels were normal, the graft was retrieved and assessed. Results: Viability of human islets was unaltered after long-distance shipment with a retrieval rate of 88.3% ± 1.9%. After encapsulation, the viability was unchanged (before encapsulation 86.1% ± 0.7% vs after encapsulation 80.8 ± 0.7%) at 11 days after isolation. Function in vitro of nonencapsulated and encapsulated islets was unaffected with a stimulation index of 2.2 and 1.9, respectively. Euglycemia was achieved in 100% mice receiving 2000 and 3000 islet equivalents of nonencapsulated and encapsulated islets, respectively. Capsules retrieved after transplantation was intact, free floating, and contained viable islets. Conclusion: Human islets can be shipped safely for long distances without compromising viability and function even after encapsulation and culture.Abbreviations: IEQs - islet equivalents, NOD/SCID - nonobese diabetic/severe combined immunodeficient


Stem Cells and Development | 2014

Hepatocyte-Like Cells Derived from Human Amniotic Epithelial Cells Can Be Encapsulated Without Loss of Viability or Function In Vitro

Vijesh Vaghjiani; Vijayaganapathy Vaithilingam; Indah Saraswati; Adnan Sali; Padma Murthi; Bill Kalionis; Bernard E. Tuch; Ursula Manuelpillai

Placenta derived human amniotic epithelial cells (hAEC) are an attractive source of stem cells for the generation of hepatocyte-like cells (HLC) for therapeutic applications to treat liver diseases. During hAEC differentiation into HLC, they become increasingly immunogenic, which may result in immune cell-mediated rejection upon transplantation into allogeneic recipients. Placing cells within devices such as alginate microcapsules can prevent immune cell-mediated rejection. The aim of this study was to investigate the characteristics of HLC generated from hAEC and to examine the effects of encapsulation on HLC viability, gene expression, and function. hAEC were differentiated for 4 weeks and evaluated for hepatocyte-specific gene expression and function. Differentiated cells were encapsulated in barium alginate microcapsules and cultured for 7 days and the effect of encapsulation on cell viability, function, and hepatocyte related gene expression was determined. Differentiated cells performed key functions of hepatocytes including urea synthesis, drug-metabolizing cytochrome P450 (CYP)3A4 activity, indocyanine green (ICG) uptake, low-density lipoprotein (LDL) uptake, and exhibited glutathione antioxidant capacity. A number of hepatocyte-related genes involved in fat, cholesterol, bile acid synthesis, and xenobiotic metabolism were also expressed showing that the hAEC had differentiated into HLC. Upon encapsulation, the HLC remained viable for at least 7 days in culture, continued to express genes involved in fat, cholesterol, bile acid, and xenobiotic metabolism and had glutathione antioxidant capacity. CYP3A4 activity and urea synthesis by the encapsulated HLC were higher than that of monolayer HLC cultures. Functional HLC can be derived from hAEC, and HLC can be encapsulated within alginate microcapsules without losing viability or function in vitro.


Current Stem Cell Research & Therapy | 2013

Deriving hepatocyte-like cells from placental cells for transplantation.

Vijesh Vaghjiani; Vijayaganapathy Vaithilingam; Bernard E. Tuch; William Sievert; Ursula Manuelpillai

Human hepatocyte transplantation is being trialled in lieu of orthotopic liver transplants for patients with acute and chronic liver diseases. Stem cells that can be differentiated into hepatocyte-like cells may replace human hepatocytes that are difficult to source, culture and in critically short supply. Hepatocyte-like cells have been derived from embryonic and adult tissue stem cells using a combination of growth factors and chemical inducers. Stem cells derived from the human placenta have gained interest due to the unlimited supply of placental tissue, minimal issues associated with stem cell retrieval from placental tissue and the large yields of stem cells that can be obtained. Placental stem cells have been characterised and differentiated into hepatocyte-like cells. This review summarises the literature relating to the differentiation of human placental stem cells into hepatocyte-like cells, the characterisation of the differentiated cells, testing the functionality of the hepatocyte-like cells in pre-clinical animal models of liver disease and biomaterials used for culturing and transplantation of these cells into extra-hepatic sites.


Cell Transplantation | 2016

Coencapsulation of Target Effector Cells with Mesenchymal Stem Cells Reduces Pericapsular Fibrosis and Improves Graft Survival in a Xenotransplanted Animal Model

Vijayaganapathy Vaithilingam; Margaret D. M. Evans; Anthony Rowe; Penelope A. Bean; Bernard E. Tuch

Pericapsular fibrotic overgrowth (PFO) is a problem that thwarts full implementation of cellular replacement therapies involving encapsulation in an immunoprotective material, such as for the treatment of diabetes. Mesenchymal stem cells (MSCs) have inherent anti-inflammatory properties. We postulated that coencapsulation of MSCs with the target cells would reduce PFO. A hepatoinsulinoma cell line (HUH7) was used to model human target cells and was coencapsulated with either human or mouse MSCs at different ratios in alginate microcapsules. Viability of encapsulated cells was assessed in vitro and xenografted either intraperitoneally or subcutaneously into C57BL/6 mice. Graft retrieval was performed at 3 weeks posttransplantation and assessed for PFO. Coencapsulation of human MSCs (hMSCs) or mouse MSCs (mMSCs) with HUH7 at different ratios did not alter cell viability in vitro. In vivo data from intraperitoneal infusions showed that PFO for HUH7 cells coencapsulated with hMSCs and mMSCs in a ratio of 1:1 was significantly reduced by ~30% and ~35%, respectively, compared to HUH7 encapsulated alone. PFO for HUH7 cells was reduced by ~51% when the ratio of mMSC/HUH7 was increased to 2:1. Implanting the microcapsules subcutaneously rather than intraperitoneally substantially reduced PFO in all treatment groups, which was most significant in the mMSC/HUH7 2:1 group with a ~53% reduction in PFO compared with HUH7 alone. Despite the reduced PFO reaction to the individual microcapsules implanted subcutaneously, all microcapsule treatment groups were contained in a vascularized fibrotic pouch at 3 weeks. The presence of MSCs in microcapsules retrieved from these fibrotic pouches improved graft survival with significantly higher cell viabilities of 83.1 ± 0.6% and 79.1 ± 0.8% seen with microcapsules containing mMSC/HUH7 at 2:1 and 1:1 ratios, respectively, compared to HUH7 alone (51.5 ± 0.7%) transplanted subcutaneously. This study showed that coencapsulation of MSCs with target cells has a dose-dependent effect on reducing PFO and improving graft survival when implanted either intraperitoneally or subcutaneously in a stringent xenotransplantation setting.


Scientific Reports | 2017

Co-encapsulation and co-transplantation of mesenchymal stem cells reduces pericapsular fibrosis and improves encapsulated islet survival and function when allografted

Vijayaganapathy Vaithilingam; Margaret D. M. Evans; Denise Lewy; Penelope A. Bean; Sumeet Bal; Bernard E. Tuch

Pericapsular fibrotic overgrowth (PFO) is associated with poor survival of encapsulated islets. A strategy to combat PFO is the use of mesenchymal stem cells (MSC). MSC have anti-inflammatory properties and their potential can be enhanced by stimulation with proinflammatory cytokines. This study investigated whether co-encapsulation or co-transplantation of MSC with encapsulated islets would reduce PFO and improve graft survival. Stimulating MSC with a cytokine cocktail of IFN-γ and TNF-α enhanced their immunosuppressive potential by increasing nitric oxide production and secreting higher levels of immunomodulatory cytokines. In vitro, co-encapsulation with MSC did not affect islet viability but significantly enhanced glucose-induced insulin secretion. In vivo, normoglycemia was achieved in 100% mice receiving islets co-encapsulated with stimulated MSC as opposed to 71.4% receiving unstimulated MSC and only 9.1% receiving encapsulated islets alone. Microcapsules retrieved from both unstimulated and stimulated MSC groups had significantly less PFO with improved islet viability and function compared to encapsulated islets alone. Levels of peritoneal immunomodulatory cytokines IL-4, IL-6, IL-10 and G-CSF were significantly higher in MSC co-encapsulated groups. Similar results were obtained when encapsulated islets and MSC were co-transplanted. In summary, co-encapsulation or co-transplantation of MSC with encapsulated islets reduced PFO and improved the functional outcome of allotransplants.


Scientific Reports | 2017

In Vitro and In Vivo Biocompatibility Evaluation of Polyallylamine and Macromolecular Heparin Conjugates Modified Alginate Microbeads

Vijayaganapathy Vaithilingam; Bjørg Steinkjer; Liv Ryan; Rolf Larsson; Bernard E. Tuch; Jose Oberholzer; Anne Mari Rokstad

Host reactivity to biocompatible immunoisolation devices is a major challenge for cellular therapies, and a human screening model would be of great value. We designed new types of surface modified barium alginate microspheres, and evaluated their inflammatory properties using human whole blood, and the intraperitoneal response after three weeks in Wistar rats. Microspheres were modified using proprietary polyallylamine (PAV) and coupled with macromolecular heparin conjugates (Corline Heparin Conjugate, CHC). The PAV-CHC strategy resulted in uniform and stable coatings with increased anti-clot activity and low cytotoxicity. In human whole blood, PAV coating at high dose (100 µg/ml) induced elevated complement, leukocyte CD11b and inflammatory mediators, and in Wistar rats increased fibrotic overgrowth. Coating of high dose PAV with CHC significantly reduced these responses. Low dose PAV (10 µg/ml) ± CHC and unmodified alginate microbeads showed low responses. That the human whole blood inflammatory reactions paralleled the host response shows a link between inflammatory potential and initial fibrotic response. CHC possessed anti-inflammatory activity, but failed to improve overall biocompatibility. We conclude that the human whole blood assay is an efficient first-phase screening model for inflammation, and a guiding tool in development of new generation microspheres for cell encapsulation therapy.


Tissue Engineering Part A | 2014

Beneficial Effects of Coating Alginate Microcapsules with Macromolecular Heparin Conjugates–In Vitro and In Vivo Study

Vijayaganapathy Vaithilingam; Gabriela Kolláriková; Meirigeng Qi; Rolf Larsson; Igor Lacík; Kjetil Formo; Enza Marchese; Jose Oberholzer; Gilles J. Guillemin; Bernard Tuch


Archive | 2013

Bioencapsulation of Living Cells for Diverse Medical Applications

Eva Maria Brandtner; John A. Dangerfield; Jean-Pierre Abastado; Shigeki Arii; S. Bilodeau; Randolph Corteling; Thomas Czerny; Jayne L. Foster; Walter H. Günzburg; Jean-Pierre Hallé; Rosa Martin Hernández; Cornelius Kaspar; Koei Kawakami; Tania López-Méndez; Ainhoa Murua; Gorka Orive; Viktoria Ortner; José Luis Pedraz Muñoz; Brian Salmons; Sakai Shinji; John Sinden; Shinji Tanaka; Bernard E. Tuch; Vijayaganapathy Vaithilingam

Collaboration


Dive into the Vijayaganapathy Vaithilingam's collaboration.

Top Co-Authors

Avatar

Bernard E. Tuch

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Jose Oberholzer

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Margaret D. M. Evans

Commonwealth Scientific and Industrial Research Organisation

View shared research outputs
Top Co-Authors

Avatar

Penelope A. Bean

Commonwealth Scientific and Industrial Research Organisation

View shared research outputs
Top Co-Authors

Avatar

Sumeet Bal

Commonwealth Scientific and Industrial Research Organisation

View shared research outputs
Top Co-Authors

Avatar

Ursula Manuelpillai

Monash Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Vijesh Vaghjiani

Monash Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge