Displaying publications 1 - 20 of 67 in total

Abstract:
Sort:
  1. Ferdaos N, Nathan S, Nordin N
    Med J Malaysia, 2008 Jul;63 Suppl A:75-6.
    PMID: 19024991
    Amniotic fluid (AF) serves as an excellent alternative source of pluripotent stem cells, as they are not bound with ethical issues and the stem cells are more primitive than adult stem (AS) cells. Hence, they have higher potential. Here we aim to isolate and characterize pluripotent stem cells from mid-term and full-term pregnant rat amniotic fluid. The results demonstrate the evidence of heterogeneous population of cells in the amniotic fluid and some of the cells morphology shows similarity with ES cells.
    Matched MeSH terms: Pluripotent Stem Cells/cytology*
  2. Simat SF, Chua KH, Abdul Rahman H, Tan AE, Tan GC
    Med J Malaysia, 2008 Jul;63 Suppl A:53-4.
    PMID: 19024980
    The aim of the study is to evaluate the stemness gene expression of cultured human amniotic epithelial cells (HAECs) in serial passages. HAECs obtained from human term placentae were cultured in F12:DMEM(1:1) + 10% FBS +10ng/ml EGF in serial passages (P0, P1, P2 and P4). Quantitative RT-PCR was used to assess the gene expression analysis. The results showed that cultured HAECs expressed and downregulated the stemness genes expression for Oct-4, Sox-2, Nanog3, FGF4, Rex-1, FZD-9, BST-1 ABCG2. However, vimentin and nestin gene expression were upregulated. The results suggested that cultured HAECs may have pluripotent and multipotent properties.
    Matched MeSH terms: Pluripotent Stem Cells/cytology*; Pluripotent Stem Cells/transplantation
  3. Mok PL, Cheong SK, Leong CF, Othman A
    Cytotherapy, 2008;10(2):116-24.
    PMID: 18368590 DOI: 10.1080/14653240701816996
    Mesenchymal stromal cells (MSC) are pluripotent progenitor cells that can be found in human bone marrow (BM). These cells have low immunogenicity and could suppress alloreactive T-cell responses. In the current study, MSC were tested for their capacity to carry and deliver the erythropoietin (EPO) gene in vitro.
    Matched MeSH terms: Pluripotent Stem Cells/cytology
  4. Hui,J.H.P., Azura M., Lee, E.H.
    Malays Orthop J, 2009;3(1):4-12.
    MyJurnal
    Regenerative medicine using stem cell therapy has sparked much interest in this 21st century not only because of the controversies that surround the ethics involving pluripotent stem cells but their potential for use in the clinic. The ability of stem cells to repair and regenerate new tissues and organs holds tremendous promise for the treatment of many serious diseases and injuries. This review provides a brief summary of the current status of research in stem cells with special emphasis on where we are in terms of the possible clinical application of stem cell therapy in orthopaedic surgery. We look at the available evidence and examine the ethical issues and considerations associated with the clinical use of stem cells.
    Matched MeSH terms: Pluripotent Stem Cells
  5. Das AK, Pal R
    J Tissue Eng Regen Med, 2010 Aug;4(6):413-21.
    PMID: 20084623 DOI: 10.1002/term.258
    Pluripotent stem cells possess the unique property of differentiating into all other cell types of the human body. Further, the discovery of induced pluripotent stem cells (iPSCs) in 2006 has opened up new avenues in clinical medicine. In simple language, iPSCs are nothing but somatic cells reprogrammed genetically to exhibit pluripotent characteristics. This process utilizes retroviruses/lentiviruses/adenovirus/plasmids to incorporate candidate genes into somatic cells isolated from any part of the human body. It is also possible to develop disease-specific iPSCs which are most likely to revolutionize research in respect to the pathophysiology of most debilitating diseases, as these can be mimicked ex vivo in the laboratory. These models can also be used to study the safety and efficacy of known drugs or potential drug candidates for a particular diseased condition, limiting the need for animal studies and considerably reducing the time and money required to develop new drugs. Recently, functional neurons, cardiomyocytes, pancreatic islet cells, hepatocytes and retinal cells have been derived from human iPSCs, thus re-confirming the pluripotency and differentiation capacity of these cells. These findings further open up the possibility of using iPSCs in cell replacement therapy for various degenerative disorders. In this review we highlight the development of iPSCs by different methods, their biological characteristics and their prospective applications in regenerative medicine and drug screening. We further discuss some practical limitations pertaining to this technology and how they can be averted for the betterment of human life.
    Matched MeSH terms: Induced Pluripotent Stem Cells/cytology*; Induced Pluripotent Stem Cells/metabolism
  6. Govindasamy V, Abdullah AN, Ronald VS, Musa S, Ab Aziz ZA, Zain RB, et al.
    J Endod, 2010 Sep;36(9):1504-15.
    PMID: 20728718 DOI: 10.1016/j.joen.2010.05.006
    Lately, several new stem cell sources and their effective isolation have been reported that claim to have potential for therapeutic applications. However, it is not yet clear which type of stem cell sources are most potent and best for targeted therapy. Lack of understanding of nature of these cells and their lineage-specific propensity might hinder their full potential. Therefore, understanding the gene expression profile that indicates their lineage-specific proclivity is fundamental to the development of successful cell-based therapies.
    Matched MeSH terms: Pluripotent Stem Cells/cytology; Pluripotent Stem Cells/physiology
  7. Nordin N, Lai MI, Veerakumarasivam A, Ramasamy R, Abdullah S, Wendy-Yeo WY, et al.
    Med J Malaysia, 2011 Mar;66(1):4-9.
    PMID: 23765134 MyJurnal
    The development of induced pluripotent stem cells (iPSCs) has been met with much enthusiasm and hailed as a breakthrough discovery by the scientific and research communities amidst the divisive and ongoing debates surrounding human embryonic stem cells (hESC) research. The discovery reveals the fact that embryonic pluripotency can be generated from adult somatic cells by the induction of appropriate transcriptional factor genes essential for maintaining the pluripotency. They provide an alternative source for pluripotent stem cells, thus representing a powerful new research tool besides their potential application in the field of regenerative medicine. In this review, the historical background of iPSCs generation will be discussed together with their properties and characteristics as well as their potential therapeutic applications.
    Matched MeSH terms: Induced Pluripotent Stem Cells*
  8. Lai MI, Wendy-Yeo WY, Ramasamy R, Nordin N, Rosli R, Veerakumarasivam A, et al.
    J Assist Reprod Genet, 2011 Apr;28(4):291-301.
    PMID: 21384252 DOI: 10.1007/s10815-011-9552-6
    Direct reprogramming of somatic cells into induced pluripotent stem (iPS) cells has emerged as an invaluable method for generating patient-specific stem cells of any lineage without the use of embryonic materials. Following the first reported generation of iPS cells from murine fibroblasts using retroviral transduction of a defined set of transcription factors, various new strategies have been developed to improve and refine the reprogramming technology. Recent developments provide optimism that the generation of safe iPS cells without any genomic modification could be derived in the near future for the use in clinical settings. This review summarizes current and evolving strategies in the generation of iPS cells, including types of somatic cells for reprogramming, variations of reprogramming genes, reprogramming methods, and how the advancement iPS cells technology can lead to the future success of reproductive medicine.
    Matched MeSH terms: Induced Pluripotent Stem Cells/cytology*
  9. Mamidi MK, Pal R, Mori NA, Arumugam G, Thrichelvam ST, Noor PJ, et al.
    J Cell Biochem, 2011 May;112(5):1353-63.
    PMID: 21337383 DOI: 10.1002/jcb.23052
    Among the different parameters governing the successful derivation and expansion of human embryonic stem cells (hESC), feeder layers play the most important role. Human feeders in form of human mesenchymal stromal cells (hMSCs) and human foreskin fibroblasts (HFFs) lay the foundation for eradication of animal-derived hESC culture system. In this study we explored the potential of human foreskin derived mesenchymal like stromal cells (HF-MSCs) to support self renewal and pluripotency of hESC. The MSCs isolated from human foreskin were found to be resistant to standard concentrations and duration of mitomycin-C treatment. Growth pattern, gene profiling (Oct-4, Nanog, Sox-2, Rex-1), cytoskeletal protein expression (vimentin, nestin) and tri-lineage differentiation potential into adipocytes, chondrocytes and osteocytes confirmed their mesenchymal stromal cell status. Further, the HF-MSCs were positive for CD105, CD166, CD73, CD44, CD90, SSEA-4, and negative for CD34, CD45, HLA-DR cell-surface markers and were found to exhibit BM-MSC-like characteristics. hESC lines co-cultured with HF-MSC feeders showed expression of expected pluripotent transcription factors Oct-4, Nanog, Sox-2, GDF-3, Rex-1, STELLAR, ABCG2, Dppa5, hTERT; surface markers SSEA-4, TRA-1-81 and maintained their cytogenetic stability during long term passaging. These novel feeders also improved the formation of embryoid bodies (EBs) from hESC which produced cell types representing three germ layers. This culture system has the potential to aid the development of clinical-grade hESCs for regenerative medicine and drug screening. Further, we envisage foreskin can serve as a valuable source of alternative MSCs for specific therapeutic applications.
    Matched MeSH terms: Pluripotent Stem Cells/cytology*; Pluripotent Stem Cells/metabolism
  10. Mohamad Buang ML, Seng HK, Chung LH, Saim AB, Idrus RB
    Arch Med Res, 2012 Jan;43(1):83-8.
    PMID: 22374243 DOI: 10.1016/j.arcmed.2012.01.012
    BACKGROUND AND AIMS: Tissue engineering strategy has been considered as an alternative treatment for diabetes mellitus due to lack of permanent pharmaceutical treatment and islet donors for transplantation. Various cell lines have been used to generate functional insulin-producing cells (IPCs) including progenitor pancreatic cell lines, embryonic stem cells (ESCs), umbilical cord blood stem cells (UCB-SCs), adult bone marrow stem cells (BMSCs), and adipose tissue-derived stem cells (ADSCs).

    METHODS: Human ADSCs from lipoaspirated abdominal fat tissue was differentiated into IPCs following a two-step induction protocol based on a combination of alternating high and low glucose, nicotinamide, activin A and glucagon-like peptide 1 (GLP-1) for a duration of 3 weeks. During differentiation, histomorphological changes of the stem cells towards pancreatic β-islet characteristics were observed via light microscope and transmission electron microscope (TEM). Dithizone (DTZ) staining, which is selective towards IPCs, was used to stain the new islet-like cells. Production of insulin hormone by the cells was analyzed via enzyme-linked immunosorbent assay (ELISA), whereas its hormonal regulation was tested via a glucose challenge test.

    RESULTS: Histomorphological changes of the differentiated cells were noted to resemble pancreatic β-cells, whereas DTZ staining positively stained the cells. The differentiated cells significantly produced human insulin as compared to the undifferentiated ADSCs, and its production was increased with an increase of glucose concentration in the culture medium.

    CONCLUSIONS: These initial data indicate that human lipoaspirated ADSCs have the potential to differentiate into functional IPCs, and could be used as a therapy to treat diabetes mellitus in the future.

    Matched MeSH terms: Induced Pluripotent Stem Cells/physiology*; Induced Pluripotent Stem Cells/ultrastructure
  11. Teoh HK, Cheong SK
    Malays J Pathol, 2012 Jun;34(1):1-13.
    PMID: 22870592 MyJurnal
    Induced pluripotent stem cells (iPSC) are derived from human somatic cells through ectopic expression of transcription factors. This landmark discovery has been considered as a major development towards patient-specific iPSC for various biomedical applications. Unlimited self renewal capacity, pluripotency and ease of accessibility to donor tissues contribute to the versatility of iPSC. The therapeutic potential of iPSC in regenerative medicine, cell-based therapy, disease modelling and drug discovery is indeed very promising. Continuous progress in iPSC technology provides clearer understanding of disease pathogenesis and ultimately new optimism in developing treatment or cure for human diseases.
    Matched MeSH terms: Induced Pluripotent Stem Cells/cytology*; Induced Pluripotent Stem Cells/transplantation
  12. Srijaya TC, Pradeep PJ, Zain RB, Musa S, Abu Kasim NH, Govindasamy V
    Stem Cells Int, 2012;2012:423868.
    PMID: 22654919 DOI: 10.1155/2012/423868
    Induced pluripotent stem cell-based therapy for treating genetic disorders has become an interesting field of research in recent years. However, there is a paucity of information regarding the applicability of induced pluripotent stem cells in dental research. Recent advances in the use of induced pluripotent stem cells have the potential for developing disease-specific iPSC lines in vitro from patients. Indeed, this has provided a perfect cell source for disease modeling and a better understanding of genetic aberrations, pathogenicity, and drug screening. In this paper, we will summarize the recent progress of the disease-specific iPSC development for various human diseases and try to evaluate the possibility of application of iPS technology in dentistry, including its capacity for reprogramming some genetic orodental diseases. In addition to the easy availability and suitability of dental stem cells, the approach of generating patient-specific pluripotent stem cells will undoubtedly benefit patients suffering from orodental disorders.
    Matched MeSH terms: Pluripotent Stem Cells; Induced Pluripotent Stem Cells
  13. Dutta S, Singh G, Sreejith S, Mamidi MK, Husin JM, Datta I, et al.
    CNS Neurosci Ther, 2013 Jan;19(1):5-11.
    PMID: 23253099 DOI: 10.1111/cns.12027
    Neurodegenerative diseases are devastating because they cause increasing loss of cognitive and physical functions and affect an estimated 1 billion individuals worldwide. Unfortunately, no drugs are currently available to halt their progression, except a few that are largely inadequate. This mandates the search of new treatments for these progressively degenerative diseases. Neural stem cells (NSCs) have been successfully isolated, propagated, and characterized from the adult brains of mammals, including humans. The confirmation that neurogenesis occurs in the adult brain via NSCs opens up fresh avenues for treating neurological problems. The proof-of-concept studies demonstrating the neural differentiation capacity of stem cells both in vitro and in vivo have raised widespread enthusiasm toward cell-based interventions. It is anticipated that cell-based neurogenic drugs may reverse or compensate for deficits associated with neurological diseases. The increasing interest of the private sector in using human stem cells in therapeutics is evidenced by launching of several collaborative clinical research activities between Pharma giants and research institutions or small start-up companies. In this review, we discuss the major developments that have taken place in this field to position stem cells as a prospective candidate drug for the treatment of neurological disorders.
    Matched MeSH terms: Pluripotent Stem Cells/physiology
  14. Khoo TS, Hamidah Hussin N, Then SM, Jamal R
    Differentiation, 2013 Feb;85(3):110-8.
    PMID: 23722082 DOI: 10.1016/j.diff.2013.01.004
    Human embryonic stem cells (hESc) are known for its pluripotency and self renewal capability, thus possess great potential in regenerative medicine. However, the lack of suitable xenofree extracellular matrix substrate inhibits further applications or the use of hESc in cell-based therapy. In this study, we described a new differentiation method, which generates a homogeneous population of mesenchymal progenitor cells (hESc-MPC) from hESc via epithelial-mesenchymal transition. The extracellular matrix (ECM) proteins from hESc-MPC had in turn supported the undifferentiated expansion of hESc. Immunocytochemistry and flow cytometry characterization of hESc-MPC revealed the presence of early mesenchymal markers. Tandem mass spectometry analysis of ECM produced by hESc-MPC revealed the presence of a mixture of extracellular proteins which includes tenascin C, fibronectin, and vitronectin. The pluripotency of hESc (MEL-1) cultured on the ECM was maintained as shown by the expression of pluripotent genes (FoxD3, Oct-4, Tdgf1, Sox-2, Nanog, hTERT, Rex1), protein markers (SSEA-3, SSEA-4, TRA-1-81, TRA-1-60, Oct-4) and the ability to differentiate into cells representative of ectoderm, endoderm and mesoderm. In summary, we have established a xeno-free autogenic feeder free system to support undifferentiated expansion of hESc, which could be of clinical relevance.
    Matched MeSH terms: Pluripotent Stem Cells/cytology; Pluripotent Stem Cells/metabolism
  15. Daneshvar N, Abdullah R, Shamsabadi FT, How CW, Mh MA, Mehrbod P
    Cell Biol Int, 2013 May;37(5):415-9.
    PMID: 23504853 DOI: 10.1002/cbin.10051
    Nanotechnology has provided new technological opportunities, which could help in challenges confronting stem cell research. Polyamidoamine (PAMAM) dendrimers, a new class of macromolecular polymers with high molecular uniformity, narrow molecular distribution specific size and shape and highly functionalised terminal surface have been extensively explored for biomedical application. PAMAM dendrimers are also nanospherical, hyperbranched and monodispersive molecules exhibiting exclusive properties which make them potential carriers for drug and gene delivery.
    Matched MeSH terms: Induced Pluripotent Stem Cells/cytology; Induced Pluripotent Stem Cells/metabolism
  16. Safwani WK, Makpol S, Sathapan S, Chua KH
    Cell Tissue Bank, 2013 Jun;14(2):289-301.
    PMID: 22476937 DOI: 10.1007/s10561-012-9309-1
    Adipose tissue is a source of multipotent stem cells and it has the ability to differentiate into several types of cell lineages such as neuron cells, osteogenic and adipogenic cells. Most studies on human adipose-derived stem cells (ASCs) have been carried out at the early passages. For clinical usage, ASCs need to be expanded in vitro for a period of time to get sufficient cells for transplantation into patients. However, the impact of long-term culture on ASCs molecular characteristics has not been established yet. Several studies have also shown that osteogenic and adipogenic cells have the ability to switch pathways during in vitro culture as they share the same progenitor cells. This data is important to ensure their functionality and efficacy before being used clinically in the treatment of bone diseases. Therefore, we aim to investigate the effect of long-term culture on the adipogenic, stemness and osteogenic genes expression during osteogenic induction of ASCs. In this study, the molecular characteristics of ASCs during osteogenic induction in long-term culture was analysed by observing their morphological changes during induction, analysis of cell mineralization using Alizarin Red staining and gene expression changes using quantitative RT-PCR. Morphologically, cell mineralization at P20 was less compared to P5, P10 and P15. Adipogenesis was not observed as negative lipid droplets formation was recorded during induction. The quantitative PCR data showed that adipogenic genes expression e.g. LPL and AP2 decreased but PPAR-γ was increased after osteogenic induction in long-term culture. Most stemness genes decreased at P5 and P10 but showed no significant changes at P15 and P20. While most osteogenic genes increased after osteogenic induction at all passages. When compared among passages after induction, Runx showed a significant increased at P20 while BSP, OSP and ALP decreased at later passage (P15 and P20). During long-term culture, ASCs were only able to differentiate into immature osteogenic cells.
    Matched MeSH terms: Pluripotent Stem Cells/metabolism; Pluripotent Stem Cells/pathology*
  17. Romli F, Alitheen NB, Hamid M, Ismail R, Abd Rahman NM
    J Cell Biochem, 2013 Jun;114(6):1230-7.
    PMID: 23239017 DOI: 10.1002/jcb.24477
    The first successful attempt to reprogram somatic cell into embryonic-like stem cell was achieved on 2006. Since then, it had sparked a race against time to bring this wonderful invention from bench to bedside but it is not easily achieved due to severe problems in term of epigenetic and genomic. With each problem arise, new technique and protocol will be constructed to try to overcome it. This review addresses the various techniques made available to create iPSC with problems hogging down the technique.
    Matched MeSH terms: Induced Pluripotent Stem Cells/physiology*
  18. Gao L, Thilakavathy K, Nordin N
    Cell Biol Int, 2013 Sep;37(9):875-87.
    PMID: 23619972 DOI: 10.1002/cbin.10120
    At the early stages of mammalian development, a number of developmentally plastic cells appear that possess the ability to give rise to all of the differentiated cell types normally derived from the three primary germ layers - unique character known as pluripotency. To date, embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have been shown to be truly pluripotent. However, recent studies have revealed a variety of other cells that demonstrate pluripotentiality, including very small embryonic-like stem cells (VSELs), amniotic fluid stem cells (AFSCs), marrow-isolated adult multilineage inducible cells (MIAMI) and multipotent adult precursor cells (MAPCs). This review summarises key features of these six kinds of pluripotent and potentially pluripotent stem cells (ESCs, iPSCs, VSELs, AFSCs, MIAMI and MAPCs) and the evidence for their pluripotency properties.
    Matched MeSH terms: Induced Pluripotent Stem Cells/cytology*; Induced Pluripotent Stem Cells/metabolism
  19. Xin LZ, Govindasamy V, Musa S, Abu Kasim NH
    Med Hypotheses, 2013 Oct;81(4):704-6.
    PMID: 23932760 DOI: 10.1016/j.mehy.2013.07.032
    Dental tissues contains stem cells or progenitors that have high proliferative capacity, are clonogenic in vitro and demonstrate the ability to differentiate to multiple type cells involving neurons, bone, cartilage, fat and smooth muscle. Numerous experiments have demonstrated that the multipotent stem cells are not rejected by immune system and therefore it may be possible to use these cells in allogeneic settings. In addition, these remarkable cells are easily abundantly available couple with less invasive procedure in isolating comparing to bone marrow aspiration. Here we proposed dental stem cells as candidate for cardiac regeneration based on its immature characteristic and propensity towards cardiac lineage via PI3-Kinase/Aktsignalling pathway.
    Matched MeSH terms: Pluripotent Stem Cells/transplantation*
  20. Musa S, Xin LZ, Govindasamy V, Fuen FW, Kasim NH
    Expert Opin Biol Ther, 2014 Jan;14(1):63-73.
    PMID: 24191782 DOI: 10.1517/14712598.2014.858694
    Acute myocardial infarction is the primary cause of heart disease-related death in the world. Reperfusion therapy is currently the backbone of treatment for acute myocardial infarction albeit with many limitations. With the emergence of stem cells as potential therapeutic agents, attempts in using them to enhance cardiac function have increased exponentially. However, it has its own disadvantages, and we postulate that the primary drawback is choosing the right cell type and solving this may significantly contribute to ambitious goal of using stem cells in the regeneration medicine.
    Matched MeSH terms: Induced Pluripotent Stem Cells/transplantation*
Filters
Contact Us

Please provide feedback to Administrator (afdal@afpm.org.my)

External Links