Induced Pluripotent Stem Cell

Similar documents
Fundamental properties of Stem Cells

John Gurdon was testing the hypothesis of genomic equivalence or that when cells divide they retain a full genomic compliment.

hpsc Growth Medium DXF Dr. Lorna Whyte

Stem Cel s Key Words:

Molecular Medicine. Stem cell therapy Gene therapy. Immunotherapy Other therapies Vaccines. Medical genomics

ANAT 2341 Embryology Lecture 18 Stem Cells

Stem Cells & Neurological Disorders. Said Ismail Faculty of Medicine University of Jordan

ANNOUNCEMENTS. HW2 is due Thursday 2/4 by 12:00 pm. Office hours: Monday 12:50 1:20 (ECCH 134)

LIFE. How-to 2 Cloning and Epigenetics. More great student questions of the day. 1. Reproductive vs therapeutic cloning

Stem Cell Research 101

Cloning. 1. What is cloning: Natural and artificial 2. Cloning of what? 3. Embryonic development of multi-cellular organisms:

Stem cell: a cell capable of 1) tissue plasticity - make different cell types 2) infinite self renewal through asymmetric division

Xeno-Free Systems for hesc & hipsc. Facilitating the shift from Stem Cell Research to Clinical Applications

Des cellules-souches dans le poumon : pourquoi faire?

NPTEL Biotechnology Tissue Engineering. Stem cells

DB3230 Midterm 1 11/15/2013 Name:

Stem Cells: Introduction and Prospects in Regenerative Medicine.

Genetic Basis of Development & Biotechnologies

Stem Cell Services. Driving Innovation for Stem Cell Researchers

Lecture 24 Differentiation and stem cells

Directed differentiation of human embryonic stem cells

Manufacturing and Characterization Challenges for Human Stem Cell Products

Assuring Pluripotency of ESC and ipsc Lines

ANAT 3231 Cell Biology Lecture 21 Stem Cells

What Are Stem Cells? [

Embryonic development, epigenics and somatic cell nuclear transfer - The science and its social implications -

Lesson 7A Specialized Cells, Stem Cells & Cellular Differentiation

A Survey of Genetic Methods

TITLE: Induced Pluripotent Stem Cells as Potential Therapeutic Agents in NF1

BME 177. Engineering stem cells. Gayatri Pal Lecturer Biomolecular Engineering

Unit 1: DNA and the Genome. Sub-Topic (1.4) Cellular Differentiation

Stem Cell Principle -

Genetic Technologies

Prof. Fahd M. Nasr. Faculty of Sciences Lebanese University Beirut, Lebanon.

Lecture 17. Transgenics. Definition Overview Goals Production p , ,

Jedi cells patrol the mouse

Nature Biotechnology: doi: /nbt Supplementary Figure 1

Will Stem Cells Finally Deliver Without Controversy?

Which of the following comes closest to your view? no genetic engineering whatsoever should be performed on humans.

Biology 4361 Developmental Biology Lecture 4. The Genetic Core of Development

Genetics and Genomics in Medicine Chapter 9 Questions

PhysicsAndMathsTutor.com. Question Number. Answer Additional guidance Mark. 1(a) 1. reference to stem cells being {totipotent / pluripotent} ;

Cat# Product Name amounts h NANOG (RFP-Bsd) inducible particles

Stem and cell gene therapy. Lecture 13

Cloning genes into animals. Transgenic animal carries foreign gene inserted into its genome.

Immunogenicity of Stem Cells in Therapeutic Applications. MDPB-Registry meeting nov 25th Stem cell immunogenicity in therapeutic applications

Concepts and Methods in Developmental Biology

Protocol Using the Reprogramming Ecotropic Retrovirus Set: Mouse OSKM to Reprogram MEFs into ips Cells

Dec 04, 2013

Direct Reprogramming of Fibroblasts into Cardiomyocytes

Biotechnology. Chapter 20. Biology Eighth Edition Neil Campbell and Jane Reece. PowerPoint Lecture Presentations for

Chapter 8 Healthcare Biotechnology

Premade Lentiviral Particles for human ips Stem Factors

KEY Reproductive cloning Therapeutic cloning

Differentiation. Ahmed Ihab Abdelaziz MD, PhD Associate Prof. Of Molecular Medicine NewGiza University (NGU)

What are the origins of medical practice? Humans have been involved with medical biotechnology

Supplementary Data. Supplementary Methods Three-step protocol for spontaneous differentiation of mouse induced pluripotent stem (embryonic stem) cells

Edexcel (B) Biology A-level

Future of Stem Cell Engineering. Jaeseung Jeong, Ph.D Department of Bio and Brain Engineering KAIST

Regenerative Medicine and Stem Cell Therapies

The NYSCF Research Institute

Developmental Biology 3230 Exam 1 (Feb. 6) NAME

Guided teaching hours: 4 hours

Premade Lentiviral Particles for ips Stem Factors: Single vector system. For generation of induced pluripotent stem (ips) cells and other applications

SUPPLEMENTARY INFORMATION

Therapeutic Cell Replacement. Steven McLoon Department of Neuroscience University of Minnesota

Protocol Reprogramming MEFs using the Dox Inducible Reprogramming Lentivirus Set: Mouse OKSM

Contribution and Mobilization of Mesenchymal Stem Cells in a mouse model of carbon tetrachloride-induced liver fibrosis

Understanding Stem Cells The Conference 1 Basics. 1 Basics

Genetics Lecture 19 Stem Cells. Stem Cells 4/10/2012

Disease Clinical features Genotype

STEM CELL RESEARCH PRIMER

CULTURE ENGINEERING DIFFERENTIATION CHARACTERIZATION. Stem Cell Research Solutions Promotional offers

WHAT IS EMBRYONIC STEM CELLS

Stem Cells, Regenerative Medicine and cgmp (GTP)

Pluripotent Stem Cells From Research to Therapy

A"Review" " " " " " " " " " " " " " Going"Uphill:"Inducing"Pluripotency." By#Ben#Laffin#

ips cell reprogramming

Chapter 11. How Genes Are Controlled. Lectures by Edward J. Zalisko

Stem Cells: Ethics and Oversight May 24, Sidney H. Golub, Ph.D. Interim Director Sue & Bill Gross Stem Cell Research Center

Biotechnology. Professor Andrea Garrison Biology 11 Illustrations 2010 Pearson Education, Inc., unless otherwise noted

Professor Martin Pera. University of Melbourne Melbourne

CIRM - MRC Human Somatic Cell Nuclear Transfer Workshop Report June 13-14, 2010

IN THE UNITED STATES PATENT AND TRADEMARK OFFICE BEFORE THE PATENT TRIAL AND APPEAL BOARD. BioGatekeeper, Inc. Petitioner

A Novel Platform to Enable the High-Throughput Derivation and Characterization of Feeder-Free Human ipscs

The New News in Stem Cell Research Andrés Bratt-Leal, PhD 12/1/2017

National Institute of Science and Technology in. Edital Nº 15/2008 MCT/CNPq/FNDCT/CAPES/FAPEMIG/FAPERJ/FAPESP

Cloning and Epigenetics. Developmental Readout. Foundations. Human issues. Stem cells. Cloning. Axon guidance.

Developing Targeted Stem Cell Therapeutics for Cancer. Shawn Hingtgen, Ph.D. Assistant Professor UNC Eshelman School of Pharmacy May 22 nd, 2013

Current progress and prospects of induced pluripotent stem cells

Versatility and performance of Lipofectamine Stem Transfection Reagent

Human therapeutic cloning

Supplementary Figure 1.

Mid-term review. Recitation5 03/31/2014. Stem Cell Biology and Function W4193 1

Induced Stem Cells in Research and Therapy of Neuropsychiatric Disorders

Protocol Reprogramming Human Fibroblasts into ips Cells using the Stemgent Reprogramming Lentivirus Set: Human OKSM

Stem Cells. Part 1: What is a Stem Cell? STO Stem cells are unspecialized. What does this mean?

Protocol Using a Dox-Inducible Polycistronic m4f2a Lentivirus to Reprogram MEFs into ips Cells

Genomic analysis of induced pluripotent stem (ips) cells: routes to reprogramming

Andrew Burnett, PhD DeGroot Center, Sanford Health Ethics & Humanities Section, SSOM USD

Transcription:

Induced Pluripotent Stem Cell When eventually mastered, the processes involved in the obtaining, cultivating and differencing ESC into cells of clinical interest, another practical limitation should be taken into account. In spite of the fact that HSC Banks may eventually be created allowing the establishment of large collections, the ESC would have their use restricted to hystocompatible receptor. One of the alternatives to the creation of a ESC bank involves the reprogramming of the nucleus of somatic cells in the case of a patient with no donor, by the cytoplasm of an oocyte to obtain autologous hesc. This approach is also known as therapeutic cloning, since it does not want to generate a cloned individual, but only ESC for therapeutic use. The reprogramming of the somatic nucleus induced by oocyte cytoplasm resulted from the presence of different molecules, including transcription factors and other proteins. Recently, several groups reported the induction of pluripotency in human primary fibroblasts through their transduction with viral vectors expressing the OCT4, C-MYC, KLF4 and SOX2 genes. The so called induced pluripotential stem cells (ips) have the characteristic morphology of ESCs. Express pluripotential cell markers and are capable of differentiation in vitro and in tissues derived from the three embryonic layers. This way, the generation of ips from somatic cell induction by specific factors could represent an alternative to obtain of histocompatible stem cells. Preliminary results of Dr. Dimas Tadeu Covas group from Hemocentro de Ribeirão Preto (USP) show the efficiency in the lentiviral production, in the stem cell transduction (mesenchymal and endothelial progenitor) and also in the endothelial progenitor expression profile with Nanog. Thus, this group, with the subproject 5, named Gene Modification of stem cell, has as general objective the genetic change of mesenchymal stem cell and endothelial progenitor cells with lentiviral vectors carrying the stemness genes, to transform multipotent cells into pluripotent ones with greater in vitro expansion capacity. The project has the following approaches: a) To transduce MSC with 1054-CIGWS lentiviral vector carrying one of the transcription factors (Nanog, Oct3/4, Sox 2, β-catenina, Kfl4, c- myc, Esrrb, Tcl 1and Tbx 3) together with the GFP gene sort out the positive cells by flow cytometry; b) transductions will be made one at a time and afterwards four vectors at the same time will be used to induce de ips; c) Evaluation of the

changes in the gene expression profile of the transduced cells; d) Evaluation of the morphological changes of the transformed cells; e) assess the specific embryonic stem cells markers (alkaline phosphatase and/or SSEA-1 antigens) e) evaluate if the modified cells have acquired pluripotenciality. A great limitation of pre-clinical studies with ESCs is the lack of large animal models on which these test can be performed since a series of technical/ biological difficulties prevent the establishment of ESCs from embryos of those models. Keeping this in mind, the group coordinated by Dr. Lygia da Veiga Pereira of USP in São Paulo (USP-SP) through subproject 6, entitled Establishment of induced pluripotent stem cell lines (ips) in large animal models, intends to develop a methodology to establish lines of ips from large animal models specifically from non-human primates and canine. The cells generated will be used in pre-clinical studies of lesion of spinal cord in the respective animal models. For that matter, lentiviral vectors of AddGene enterprise (EUA) will be transferred to 293 cells by co-transfection with packing vectors. Cultures of dogs and monkeys will be transduced with lentivirus expressing the reporter gene GFP for evaluation of transduction efficiency with different packing systems; fibroblast transduction animals with induction vectors according to conditions established above, and isolation of ips in culture medium specific for ESC (in case we are not able to establish the ips by using vectors with human genes of induction, the homologous of each species will be isolated by RT-PCR from embryos, pre-implantation and new species-specific vectors will be constructed and used for induction); characterization of pluripotentiality of ips by immunofluorescence, in vitro differentiation in embryonic bodies and in vivo by the measurement of the formation of teratomas; neural differentiation of animal ips by culture in media with retinoic acid; transplant of differentiated cells in spine cord lesion models. Despite the fact that is inadequate for clinical use, the ips are an important tool of basic research, mainly for cells obtained from individuals with genetic diseases Thus, the group headed by Dr. Lygia da Veiga Pereira, through subproject 7, entitled Establishment of induced pluripotent stem cell lines (ips) from fibroblasts of patients with Mendelian and multifactorial genetic diseases (with genetic component), intends to implement the methodology of generation of human ips so that it is possible to establish ips from different tissues of patients with genetic diseases of interest to the group, particularly patients who have

osseous dysplasia, type 1 diabetes, multiple sclerosis and acute promyielocytic leukemia. The ips established will be used as experimental model for the study of basic mechanisms behind the respective diseases. Furthermore, new vectors based on adenovirus will be constructed, which will not integrate to the genome, so nonmodified ips will be generated, which is better suited for clinical use. For that purpose, the following experimental approaches will be used: lentiviral vectors of AddGene enterprise (EUA) will be inserted into 293 cells by transient cotransfection with vectors; frozen mesenchymal cells of the bone marrow of patients who have type 1 diabetes and leukemia PMA cells will be expanded in culture for viral transduction; standardization of line establishment of adequate cells for induction from human peripheral blood; transduction of human cells with lentiviral vectors and selection of ips cells in culture medium for ESCs; postimmunofluorescence characterization of ips and FACS using pluripotent cell markers (OCT4, NANOG, SSEA-1,2,3,4); differentiation into embryoid bodies and characterization by immunofluorescence; teratoma formation in SCID mice; characterization of pluripotency of ips through immunofluorescence, perform differentiation into embryoid bodies in vitro, and teratoma formation assays in vivo. Although techniques for the induction of pluripotency involving the incorporation of transcription factors in the genome of somatic cells (Takahashi et al., 2006) constitute a possible approach to autologous cell therapy, it is important to stress that the practical application of cells induced to differentiation by genetic modification still has to be studied further and evaluated (Liu, 2008). On the other hand, the technique of nuclear transfer (NT) is well-established and has been capable of producing healthy animals, confirming the efficient capacity of reprogramming of a differentiated cell (Wilmut et al., 1997; Keefer et al., 2000). Additionally, when used as a receptor, the cytoplastic bovine cell has been capable of reprogramming differentiated cells from various species (Chen et al., 2002, Ilmensee et al., 2006), supporting the initial view that embryonary development is necessary for the isolation of embryonary stem cells. Together with the efficient reprogramming of the cytoplasm, the abundance and the ease in obtaining biological material makes the bovine the perfect model for the establishment of efficient methods for obtaining pluripotent cells of embryonic origin through the

reprogramming of differentiated somatic cells of various species (Cibelli et al, 1998). By keeping that in sight, the group coordinated by Dr. Flavio Vieira Meirelles (from now on called group) of USP in Pirassununga (USP) through subproject 8, entitled Autologous pluripotent cells generated from differentiated somatic cells, will have as a general goal to characterize methodologies to obtain pluri/totipotent cells from somatic cells, in term of morphology, genetic expression and epigenetic studies. For that matter, a bovine somatic cell line derived from the in vitro cultivation of adult fibroblasts will be initially established. Part of these cells will be used in the lentiviral transduction of transcription factors responsible for cellular dedifferentiation (Takahashi et al., 2006). Such cells will be characterized in the bovine model and, along with the non-modified cells; they will be used as nuclei donors in the process of nuclear transfer. Embryonary cells derived from those processes will be compared among themselves and with those somatic undifferentiated cells in terms of morphology as well as genetic expression and epigenetic expression. They will also be compared to the embryonary cells obtained by the natural fertilization processes. The one methodology that provides characteristics closes to the ones of the control group will be considered the most appropriate for the production of stem cells from differentiated somatic cells and, so, it will be utilized in the interspecific model where pluripotency cells of primates will be produced. The reprogramming of the nucleus of somatic cells by the cytoplasm of an oocyte implies in the combination between the nuclear genome of an individual and the mitochondrial of another. Considering the great potential of this kind of approach in the development of the future of cell therapy, the control of the inherited mitochondrial is very important to make possible i) the production of human stem cells by the reprogramming of the nucleus donor cell in non-human cytoplasm and, ii) the production of autologous human stem cells (Illmensse et al., 2006). For that matter, the bovine model is interesting because of the abundance of material available and for the great quantity of available knowledge on the mechanisms that regulate the mitochondrial DNA (mtdna) during embryogenesis. In bovine embryos produced by nucleus transfer of somatic cells, the percentage of mtdna of the nucleus donor cell rises between the third and fourth cellular cycle in relation to the mtdna that comes from the oocyte (Ferreira et al., 2007). On the other hand,

the centrifugation of bovine zygotes makes the mechanical depletion of part of the mitochondria possible without compromising the embryonic development because the embryo is capable of replacing the same content of mtdna observed in undepleted blastocysts (Chiaratti et al., 2008). Furthermore, through mitochondrial depletion it is possible to introduce a greater quantity of exogenous mitochondrial zygotes (Ferreira et al., submitted). After what has been exposed, the group coordinated by Dr. Flavio Vieira Meirelles (from now on called group) of USP in Pirassununga (USP) through subproject 9, entitled Animal models to study mitochondrial inheritance intra- and interspecies, will have as a general goal, to evaluate the viability of production of embryos having mtdna of inner and interspecific somatic origin and to increase the percentage of such mtdna inherited by the blastocysts. More specifically, they intent to produce bovine blastocysts (Bos taurus) that present mtdna from inner or interspecific somatic origin (B. indicus or Homo sapiens, respectively), in heteroplasmia with mtdna from embryonic origin (inherited from the oocyte) and; additionally develop a method to raise the inherited percentage in blastocysts of somatic mtdna (B. indicus or H. sapiens). For that, the following technological approaches will be used the establishment of mesenchymal cell lines originated from the B. indicus and H. sapiens; enucleation of mesenchymal cells by centrifugation and cytoplasm fusion for utilization as a cytoplasm donor (Marchington et al., 1999; Shay et al., 1975); in vitro production of bovine zygotes (B. taurus) parthenogenetic oocytes aspirated from ovaries collected on the slaughterhouse and matured in vitro (Méo et al., 2007); centrifugation of zygote to concentrate the mitochondria in one of the poles of the embryo and the removal of part of the mitochondrias through micromanipulation (Ferreira et al., submitted); cytoplasts fusion (B. indicus or H. sapiens) to deplete zygote (B. taurus) and in vitro cultivation (Inoue et al., 2000); determination of the percentage of mtdna (somatic mtdna in relation to the total quantity of mtdna) by PCR in real time immediately after fusion, at 72 hours (embryos with 5 or more cells) and at 168 hours (blastocysts) after parthenogenic activation (Ferreira et al. submitted; Ferreira et al., 2007). The percentage of mtdna will be analyzed considering the following as effect - the cytoplasm used (B. indicus and H. sapiens), the moment of the analysis (0, 72 and 168 hours) and the interaction both factors.