• Keine Ergebnisse gefunden

Comparison of in vitro cultures of astrocytes from different sources

4.5 A STROCYTES FOR IN VITRO TESTING

4.5.3 Comparison of in vitro cultures of astrocytes from different sources

Many subtypes of astrocytes are known. Unfortunately for in vitro astrocyte research, most of them are distinguished only by brain regions (Reichenbach and Wolburg 2005). Little is known about the differential expression of marker sets or about different functionalities of the diverse astrocytes subtypes in vivo. Further research into the phenotypic and functional differences of astrocytes in different regions of the brain is needed to allow subtype specification in in vitro derived astrocytes. Recently, Krencik et al. succeeded in differentiating human ESC into neural epithelial progenitor cells. These neural epithelial progenitors were then differentiated to progenitor cells in the presence of the patterning factors FGF8 (anterior) or retinoic acid (posterior). After

in stem cell-derived astrocytes. The exact subtype identity of our MEDA cultures remains to be elucidated.

Figure 4-4: Methods to derive astrocytes from embryonic stem cells developed in this thesis.

Two methods to derive functional astrocytes from murine ESC were developed in this thesis. (A) Mouse embryonic stem cell derived astrocytes (MEDA), generating GFAP-positive and GFAP-negative astrocytes, were differentiated according to a two-step protocol. In the first step, neural induction was triggered in suspension cultures for 21 days. Afterwards, the neural cell-containing aggregates were further differentiated adherently to astrocytes. The protocol generated also GFAP-negative astrocytes which, for the first time, allowed the study of functional properties of GFAP-negative astrocytes in vitro.

(B) The second protocol relied on a different strategy: First, neural progenitor cells (NPC) were derived from embryonic stem cells. These NPC can be expanded in vitro and cryopreserved or induced to differentiate into astrocytes within 3-5 days, generating pure populations of non-dividing, GFAP-positive astrocytes. gel: gelatine; PorL: poly-L-ornithin/laminin; A: no cytokines; B: EGF/bFGF; C:

CNTF/BMP4.

It is important to compare stem cell-derived astrocytes with primary isolated astrocytes, being the “gold standard” in the field. MEDA shared many of the tested functional aspects such as inflammatory activation and metabolic competence with primary astrocytes, however their staining pattern was different. Similar to primary astrocytes, the ability of the MEDA cultures to respond to CCM, and to remain quiescent without stimulation, was retained upon replating or thawing after cryopreservation (Crocker et al. 2008; Falsig et al. 2004; Henn et al. 2011).

disease, however, as mentioned before, they most likely represent a subpopulation of all astrocytes capable to grow in culture conditions they are isolated with. Primary astrocytes proliferate in vitro. MEDA obtained by the first protocol proliferate and also express nestin. This is indicative of an either activated or immature phenotype (or both). Astrocytes derived from ESC via NPC neither proliferated nor expressed nestin, which indicates a more mature phenotype. Nevertheless, the high levels of GFAP expression in these cells indicates an either immature or an activated phenotype. In vivo, high expression of GFAP is associated with astrogliosis. However as of now, this is only speculation and requires further analysis. We initiated a project that compares the mRNA expression levels in astrocytes from various sources to gain some insight into the degree of maturity of in vitro astrocytes and also to identify new suitable astrocyte markers (Götz et al., personal communication).

4.5.4 Inflammation in neurotoxicity and brain disease

Astrocytes are implicated in degenerative processes in the brain. Activation of astrocytes in a diseased brain or by exogenous substances can lead to (chronic) inflammation. An overshooting activation can result in a strong immune response including cytokine secretion ROS formation (Figure 4-5) (Dong and Benveniste 2001).

This process called astrogliosis is usually observed in patients with chronic brain diseases like AD or PD (Carpentier et al. 2008; Falsig et al. 2008).

Apart from inflammation in disease, astrocytes can also become activated upon exposure to chemicals. For example, trimethyltin derivatives induce astrogliosis in astrocytes which leads to neurotoxicity in vitro (Cristòfol et al. 2004; Holden and Coleman 2007).

Insights on a molecular detail of the contribution of astrocytes to the development of degenerative diseases have been gained in studies of the fragile X

generation of amyotrophic laterals sclerosis (ALS). This disease is characterised by a progressive loss of peripheral motor neurons. Astrocytes carrying a mutated form of superoxide dismutase (SOD1) that has been linked to ALS before (Rosen et al. 1991), were shown to secrete soluble factors that selectively kill motor neurons in co-cultures, thereby causing the disease (Di Giorgio et al. 2008; Nagai et al. 2007).

In cases where astrocytes contribute to neurotoxicity or to complex degenerative diseases such as ALS, AD or PD, standard in vitro neuronal cultures will fail as a model, because the complex interaction of the two (or more) cell types can not be studied in detail. Assays that detect neurotoxicity using co-cultures of neurons and astrocytes have already been developed (Anderl et al. 2009; Woehrling et al. 2007) and report a generally higher resistance to toxicants of neuron, when co-cultured with astrocytes. Also, as stated before, the presence of contaminating microglia, as often described for primary astrocyte preparations (Saura 2007), would hamper functional studies as microglia can produce similar inflammatory mediators as astrocytes .

Our microglia free MEDA were developed in order to use them in co-culture models to study aspects of brain inflammation and the development of neurotoxicity (Figure 4-5). Our DNT-test system based on neurons differentiated from mESC (Chapter 3.2) also contained “contaminating” astrocytes, and therefore astrocyte-mediated influences on toxicity or disease generation might also be picked up in these cultures. However, the few astrocytes were restricted to small astrocytic “islands” in the culture well and were not evenly distributed. The generation of defined co-cultures of neurons and astrocytes is therefore preferable and would yield more consistent results when studying the interplay of astrocytes and neurons in co-cultures.

4.5.5 Astrocyte metabolism: the double edged sword

Everything comes at a price. In the healthy brain, the metabolic activity of astrocytes for maintaining physiological conditions is crucial for a functioning brain.

Occasionally however, the metabolic activity of astrocytes can result in toxic metabolites. We used a substance commonly used for the induction of a Parkinson disease phenotype in mice as a model substance to demonstrate the metabolic functions of our astrocytes. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is a

enzyme monoamineoxidase B (MaoB) into 1-methyl-4-phenyl-1,2-dihydropyridinium (MPDP+) which spontaneously forms the potent neurotoxin 1-methyl-4-phenyl-pyridinium (MPP+). Using ESI-MS we showed that astrocytes differentiated from ESC converted MPTP and released MPDP+ and MPP+ into the supernatant, underlining their suitability for toxicological studies (Chapter 3.3 and 3.4). Similar data has also previously been reported for primary astrocytes (Schildknecht et al. 2009).

Figure 4-5: Astrocytes in neurotoxicity and developmental neurotoxicity.

The involvement of glial cells (astrocytes and microglia) in the generation of indirect neurotoxicity is indicated. Astrocyte activation can lead to an overshooting immune response that can lead to neuronal damage or cell death. Soluble factors and reactive oxygen species secreted by activated glial cells are

neurotoxicity testing. If omitted, substances that are converted into toxins by astrocytes will be overlooked in neuronal cultures devoid of astrocytes. Besides, their metabolising activity is important for detoxification of neurotoxins (e.g. removal of excess extracellular glutamate), and testing in cultures devoid of astrocytes could lead to hazard overestimation, because the substance might not be eliminated by metabolic activity or their concentration would not be reduced to non-cytotoxic levels in absence

of astrocytes, as it would be the case in vivo. In vitro neurotoxicity assays based on astrocyte-neuronal co-cultures should complement conventional testing with neuron mono-cultures, to guarantee a better safety assessment of potential toxicants.

I have demonstrated that astrocytes derived from stem cells can serve as a substitute for primary isolated astrocytes. They behaved similar in all functional characteristics tested and, using the protocols I developed, can be obtained in large amounts under chemically defined conditions. Furthermore, they can be cryopreserved without loss of functionality, which makes the preparation of large batches possible, which makes them ideal candidates for next generation assay development.

The future challenge is now to transfer the methods I developed to human ESC (hESC) and to delineate functional astrocytes. In human development, astrocytes do not appear before the 3rd months of development. In vitro generation of astrocytes from hESC can therefore be expected to take longer than from the mouse (Liu and Zhang 2011). In a very promising approach, Krencik et al. recently differentiated hESC to neuroepithelium and then into astroglial progenitor cells. These progenitor cells were expanded in suspension cultures as aggregates and were differentiated to astrocytes using CNTF and LIF or serum. Apart from phenotypic marker expression, these hESC-derived astrocytes also showed functional characteristics of astrocytes (Krencik et al.

2011; Liu and Zhang 2011). However, the differentiation phase required over 180 days, which raises doubts as to their practical applicability in the near future.

5 References

Abeyta MJ, Clark AT, Rodriguez RT, Bodnar MS, Pera RA, Firpo MT. 2004. Unique gene expression signatures of independently-derived human embryonic stem cell lines. Hum Mol Genet 13(6):601-8.

Abranches E, Silva M, Pradier L, Schulz H, Hummel O, Henrique D, Bekman E. 2009. Neural differentiation of embryonic stem cells in vitro: a road map to neurogenesis in the embryo.

PLoS One 4(7):e6286.

Aiba K, Sharov AA, Carter MG, Foroni C, Vescovi AL, Ko MS. 2006. Defining a developmental path to neural fate by global expression profiling of mouse embryonic stem cells and adult neural stem/progenitor cells. Stem Cells 24(4):889-95.

Allen NJ, Barres BA. 2009. Neuroscience: Glia - more than just brain glue. Nature 457(7230):675-7.

Aloisi F, Carè A, Borsellino G, Gallo P, Rosa S, Bassani A, Cabibbo A, Testa U, Levi G, Peschle C.

1992. Production of hemolymphopoietic cytokines (IL-6, IL-8, colony-stimulating factors) by normal human astrocytes in response to IL-1 beta and tumor necrosis factor-alpha. J Immunol 149(7):2358-66.

Ambrosini E, Columba-Cabezas S, Serafini B, Muscella A, Aloisi F. 2003. Astrocytes are the major intracerebral source of macrophage inflammatory protein-3alpha/CCL20 in relapsing experimental autoimmune encephalomyelitis and in vitro. Glia 41(3):290-300.

Anderl JL, Redpath S, Ball AJ. 2009. A neuronal and astrocyte co-culture assay for high content analysis of neurotoxicity. J Vis Exp 27:e1173, doi: 10.3791/1173.

Armstrong L, Saretzki G, Peters H, Wappler I, Evans J, Hole N, von Zglinicki T, Lako M. 2005.

Overexpression of telomerase confers growth advantage, stress resistance, and enhanced differentiation of ESCs toward the hematopoietic lineage. Stem Cells 23(4):516-29.

Aschner M, Kimelberg HK. 1991. The use of astrocytes in culture as model systems for evaluating neurotoxic-induced-injury. Neurotoxicology 12(3):505-17.

Assou S, Le Carrour T, Tondeur S, Ström S, A. G, Marty S, Nadal L, Pantesco V, Réme T, Hugnot JP and others. 2007. A meta-analysis of human embryonic stem cells transcriptome integrated into a web-based expression atlas. Stem Cells 25(4):961-73.

Bahramsoltani M, Plendl J, Janczyk P, Custodis P, Kaessmeyer S. 2009. Quantitation of angiogenesis and antiangiogenesis in vivo, ex vivo and in vitro - an overview. ALTEX 26(2):95-107.

Bain G, Ray WJ, Yao M, Gottlieb DI. 1996. Retinoic acid promotes neural and represses mesodermal gene expression in mouse embryonic stem cells in culture. Biochem Biophys Res Commun 223(3):691-4.

Bajramović JJ, Bsibsi M, Geutskens SB, Hassankhan R, Verhulst KC, Stege GJ, de Groot CJ, van Noort JM. 2000. Differential expression of stress proteins in human adult astrocytes in response to cytokines. J Neuroimmunol 106(1-2):14-22.

Baker M. 2010. iPS cells: potent stuff. Nat Methods 7(1):17-9.

Bal-Price AK, Hogberg HT, Buzanska L, Lenas P, van Vliet E, Hartung T. 2009. In vitro developmental neurotoxicity (DNT) testing: Relevant models and endpoints. NeuroToxicology.

Barberi T, Klivenyi P, Calingasan NY, Lee H, Kawamata H, Loonam K, Perrier AL, Bruses J, Rubio ME, Topf N and others. 2003. Neural subtype specification of fertilization and nuclear transfer embryonic stem cells and application in parkinsonian mice. Nat Biotech 21(10):1200-1207.

Battiste J, Helms AW, Kim EJ, Savage TK, Lagace DC, Mandyam CD, Eisch AJ, Miyoshi G, Johnson JE. 2007. Ascl1 defines sequentially generated lineage-restricted neuronal and oligodendrocyte precursor cells in the spinal cord. Development 134(2):285-93.

Bauer H, Bauer H-C, Haseloff RF, Blasig IE. 2005. The role of Glia in the Formation and Function of

Amit M and others. 2004. Gene expression in human embryonic stem cell lines: unique molecular signature. Blood 103(9):2956-64.

Bhattacharya B, Puri S, Puri RK. 2009. A review of gene expression profiling of human embryonic stem cell lines and their differentiated progeny. Curr Stem Cell Res Ther 4(2):98-106.

Bignami A, Eng LF, Dahl D, Uyeda CT. 1972. Localization of the glial fibrillary acidic protein in astrocytes by immunofluorescence. Brain Res 43(2):429-35.

Blomgren K, Leist M, Groc L. 2007. Pathological apoptosis in the developing brain. Apoptosis 12(5):993-1010.

Blomme EA, Yang Y, Waring JF. 2009. Use of toxicogenomics to understand mechanisms of drug-induced hepatotoxicity during drug discovery and development. Toxicol Lett 186(1):22-31.

Bortvin A, Eggan K, Skaletsky H, Akutsu H, Berry DL, Yanagimachi R, Page DC, Jaenisch R. 2003.

Incomplete reactivation of Oct4-related genes in mouse embryos cloned from somatic nuclei.

Development 130(8):1673-80.

Bottini AA, Amcoff P, Hartung T. 2007. Food for thought ... on globalisation of alternative methods.

ALTEX 24(4):255-69.

Bottini AA, Hartung T. 2009. Food for thought... on the economics of animal testing. ALTEX 26(1):3-16.

Bouhon IA, Joannides A, Kato H, Chandran S, Allen ND. 2006. Embryonic stem cell-derived neural progenitors display temporal restriction to neural patterning. Stem Cells 24(8):1908-13.

Breier JM, Gassmann K, Kayser R, Stegeman H, De Groot D, Fritsche E, Shafer TJ. 2009. Neural progenitor cells as models for high-throughput screens of developmental neurotoxicity: State of the science. Neurotoxicol Teratol 32(1):4-15.

Breier JM, Radio NM, Mundy WR, Shafer TJ. 2008. Development of a high-throughput screening assay for chemical effects on proliferation and viability of immortalized human neural progenitor cells. Toxicol Sci 105(1):119-33.

Brüstle O, Jones KN, Learish RD, Karram K, Choudhary K, Wiestler OD, Duncan ID, McKay RD.

1999. Embryonic stem cell-derived glial precursors: a source of myelinating transplants.

Science 285(5428):754-6.

Buckley NJ, Johnson R, Sun YM, Stanton LW. 2009. Is REST a regulator of pluripotency? Nature 457(7233):E5-6.

Buesen R, Genschow E, Slawik B, Visan A, Spielmann H, Luch A, Seiler A. 2009. Embryonic stem cell test remastered: comparison between the validated EST and the new molecular FACS-EST for assessing developmental toxicity in vitro. Toxicol Sci 108(2):389-400.

Buffo A, Rite I, Tripathi P, Lepier A, Colak D, Horn AP, Mori T, Götz M. 2008. Origin and progeny of reactive gliosis: A source of multipotent cells in the injured brain. Proc Natl Acad Sci U S A 105(9):3581-6.

Burette A, Jalenques I, Romand R. 1998. Developmental distribution of astrocytic proteins in the rat cochlear nucleus. Brain Res Dev Brain Res 107(2):179-89.

Buzanska L, Sypecka J, Nerini-Molteni S, Compagnoni A, Hogberg HT, del Torchio R, Domanska-Janik K, Zimmer J, Coecke S. 2009. A human stem cell-based model for identifying adverse effects of organic and inorganic chemicals on the developing nervous system. Stem Cells 27(10):2591-601.

Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, Christopherson KS, Xing Y, Lubischer JL, Krieg PA, Krupenko SA and others. 2008. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J Neurosci 28(1):264-78.

Canzonetta C, Mulligan C, Deutsch S, Ruf S, O'Doherty A, Lyle R, Borel C, Lin-Marq N, Delom F, Groet J and others. 2008. DYRK1A-dosage imbalance perturbs NRSF/REST levels, deregulating pluripotency and embryonic stem cell fate in Down syndrome. Am J Hum Genet 83(3):388-400.

Carey BW, Markoulaki S, Beard C, Hanna J, Jaenisch R. 2010. Single-gene transgenic mouse strains for reprogramming adult somatic cells. Nat Methods 7(1):56-9.

Carlone DL, Lee JH, Young SR, Dobrota E, Butler JS, Ruiz J, Skalnik DG. 2005. Reduced genomic cytosine methylation and defective cellular differentiation in embryonic stem cells lacking CpG binding protein. Mol Cell Biol 25(12):4881-91.

Carpentier PA, Duncan DS, Miller SD. 2008. Glial toll-like receptor signaling in central nervous system infection and autoimmunity. Brain Behav Immun 22(2):140-7.

136(14):2311-22.

Chapin RE, Stedman DB. 2009. Endless possibilities: stem cells and the vision for toxicology testing in the 21st century. Toxicol Sci 112(1):17-22.

Chaudhry FA, Lehre KP, van Lookeren Campagne M, Ottersen OP, Danbolt NC, Storm-Mathisen J.

1995. Glutamate transporters in glial plasma membranes: highly differentiated localizations revealed by quantitative ultrastructural immunocytochemistry. Neuron 15(3):711-20.

Chiba K, Trevor A, Castagnoli NJ. 1984. Metabolism of the neurotoxic tertiary amine, MPTP, by brain monoamine oxidase. Biochem Biophys Res Commun 120(2):574-8.

Coecke S, Goldberg AM, Allen S, Buzanska L, Calamandrei G, Crofton K, Hareng L, Hartung T, Knaut H, Honegger P and others. 2007. Workgroup report: incorporating in vitro alternative methods for developmental neurotoxicity into international hazard and risk assessment strategies.

Environ Health Perspect 115(6):924-31.

Coles JA, Deitmer JW. 2005. Extracellular Potassium and pH: Homeostasis and Signaling. In:

Kettenmann H, Ransom B, editors. Neuroglia Second Edition: Oxford University Press Inc. p 334-345.

Collins FS, Gray GM, Bucher JR. 2008. TOXICOLOGY: Transforming Environmental Health Protection. Science 319(5865):906-907.

Compston A, Coles A. 2002. Multiple sclerosis. Lancet 359(9313):1221-31.

Conti L, Cattaneo E. 2010. Neural stem cell systems: physiological players or in vitro entities? Nat Rev Neurosci 11(3):176-87.

Conti L, Pollard SM, Gorba T, Reitano E, Toselli M, Biella G, Sun Y, Sanzone S, Ying QL, Cattaneo E and others. 2005. Niche-independent symmetrical self-renewal of a mammalian tissue stem cell. PLoS Biol 3(9):e283.

Cory-Slechta DA. 1990. Lead exposure during advanced age: alterations in kinetics and biochemical effects. Toxicol Appl Pharmacol 104(1):67-78.

Coufal NG, Garcia-Perez JL, Peng GE, Yeo GW, Mu Y, Lovci MT, Morell M, O'Shea KS, Moran JV, Gage FH. 2009. L1 retrotransposition in human neural progenitor cells. . Nature 460(7259):1127-31.

Cristòfol RM, Gassó S, Vílchez D, Pertusa M, Rodríguez-Farré E, Sanfeliu C. 2004. Neurotoxic effects of trimethyltin and triethyltin on human fetal neuron and astrocyte cultures: a comparative study with rat neuronal cultures and human cell lines. Toxicol Lett 152(1):35-46.

Crocker SJ, Frausto RF, Whitton JL, Milner R. 2008. A novel method to establish microglia-free astrocyte cultures: comparison of matrix metalloproteinase expression profiles in pure cultures of astrocytes and microglia. Glia 56(11):1187-98.

Dahl D, Rueger DC, Bignami A, Weber K, Osborn M. 1981. Vimentin, the 57 000 molecular weight protein of fibroblast filaments, is the major cytoskeletal component in immature glia. . Eur J Cell Biol 24(2):191-6.

Davies JE, Pröschel C, Zhang N, Noble M, Mayer-Pröschel M, Davies SJ. 2008. Transplanted astrocytes derived from BMP- or CNTF-treated glial-restricted precursors have opposite effects on recovery and allodynia after spinal cord injury. J Biol Chem 7(7):24.

Der SD, Zhou A, Williams BR, Silverman RH. 1998. Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proc Natl Acad Sci U S A 95(26):15623-8.

Di Giorgio FP, Boulting GL, Bobrowicz S, Eggan KC. 2008. Human embryonic stem cell-derived motor neurons are sensitive to the toxic effect of glial cells carrying an ALS-causing mutation.

Cell Stem Cell 3(6):637-48.

Di Giorgio FP, Carrasco MA, Siao MC, Maniatis T, Eggan K. 2007. Non-cell autonomous effect of glia

Dziennis S, Yang D, Cheng J, Anderson KA, Alkayed NJ, Hurn PD, Lein PJ. 2008. Developmental exposure to polychlorinated biphenyls influences stroke outcome in adult rats. Environ Health Perspect 116(4):474-80.

Eckfeldt CE, Mendenhall EM, Verfaillie CM. 2005. The molecular repertoire of the 'almighty' stem cell.

Nat Rev Mol Cell Biol 6(9):726-37.

Efroni S, Melcer S, Nissim-Rafinia M, Meshorer E. 2009. Stem cells do play with dice: a statistical physics view of transcription. Cell Cycle 8(1):43-8.

Elkabetz Y, Panagiotakos G, Al Shamy G, Socci ND, Tabar V, Studer L. 2008. Human ES cell-derived neural rosettes reveal a functionally distinct early neural stem cell stage. Genes Dev 22(2):152-65.

Ema M, Mori D, Niwa H, Hasegawa Y, Yamanaka Y, Hitoshi S, Mimura J, Kawabe Y, Hosoya T, Morita M and others. 2008. Kruppel-like factor 5 is essential for blastocyst development and the normal self-renewal of mouse ESCs. Cell Stem Cell 3(5):555-67.

Eng LF, Ghirnikar RS, Lee YL. 2000. Glial fibrillary acidic protein: GFAP-thirty-one years (1969-2000). Neurochem Res 25(9-10):1439-51.

Eto K, Oyanagi S, Itai Y, Tokunaga H, Takizawa Y, Suda I. 1992. A fetal type of Minamata disease. An autopsy case report with special reference to the nervous system. Mol Chem Neuropathol 16(1-2):171-86.

Evans MJ, Kaufman MH. 1981. Establishment in culture of pluripotential cells from mouse embryos.

Nature 292(5819):154-6.

Falsig J, Latta M, Leist M. 2004. Defined inflammatory states in astrocyte cultures: correlation with susceptibility towards CD95-driven apoptosis. J Neurochem 88(1):181-93.

Falsig J, Pörzgen P, Lund S, Schrattenholz A, Leist M. 2006a. The inflammatory transcriptome of reactive murine astrocytes and implications for their innate immune function. J Neurochem 96(3):893-907.

Falsig J, Pörzgen P, Lund S, Schrattenholz A, Leist M. 2006b. The inflammatory transcriptome of reactive murine astrocytes and implications for their innate immune function. . J Neurochem 96:893-907.

Falsig J, van Beek J, Hermann C, Leist M. 2008. Molecular basis for detection of invading pathogens in the brain. J Neurosci Res 86(7):1434-47.

Farthing CR, Ficz G, Ng RK, Chan CF, Andrews S, Dean W, Hemberger M, Reik W. 2008. Global mapping of DNA methylation in mouse promoters reveals epigenetic reprogramming of pluripotency genes. PLoS Genet 4(6):e1000116.

Fatemi SH, Folsom TD, Reutiman TJ, Lee S. 2008. Expression of astrocytic markers aquaporin 4 and connexin 43 is altered in brains of subjects with autism. Synapse 62(7):501-7.

Feng B, Jiang J, Kraus P, Ng JH, Heng JC, Chan YS, Yaw LP, Zhang W, Loh YH, Han J and others.

2009. Reprogramming of fibroblasts into induced pluripotent stem cells with orphan nuclear receptor Esrrb. Nat Cell Biol 11(2):197-203.

Feng L, Hatten ME, Heintz N. 1994. Brain lipid-binding protein (BLBP): a novel signaling system in

Feng L, Hatten ME, Heintz N. 1994. Brain lipid-binding protein (BLBP): a novel signaling system in