Network


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

Hotspot


Dive into the research topics where Ernesto Lujan is active.

Publication


Featured researches published by Ernesto Lujan.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Direct conversion of mouse fibroblasts to self-renewing, tripotent neural precursor cells

Ernesto Lujan; Soham Chanda; Henrik Ahlenius; Thomas C. Südhof; Marius Wernig

We recently showed that defined sets of transcription factors are sufficient to convert mouse and human fibroblasts directly into cells resembling functional neurons, referred to as “induced neuronal” (iN) cells. For some applications however, it would be desirable to convert fibroblasts into proliferative neural precursor cells (NPCs) instead of neurons. We hypothesized that NPC-like cells may be induced using the same principal approach used for generating iN cells. Toward this goal, we infected mouse embryonic fibroblasts derived from Sox2-EGFP mice with a set of 11 transcription factors highly expressed in NPCs. Twenty-four days after transgene induction, Sox2-EGFP+ colonies emerged that expressed NPC-specific genes and differentiated into neuronal and astrocytic cells. Using stepwise elimination, we found that Sox2 and FoxG1 are capable of generating clonal self-renewing, bipotent induced NPCs that gave rise to astrocytes and functional neurons. When we added the Pou and Homeobox domain-containing transcription factor Brn2 to Sox2 and FoxG1, we were able to induce tripotent NPCs that could be differentiated not only into neurons and astrocytes but also into oligodendrocytes. The transcription factors FoxG1 and Brn2 alone also were capable of inducing NPC-like cells; however, these cells generated less mature neurons, although they did produce astrocytes and even oligodendrocytes capable of integration into dysmyelinated Shiverer brain. Our data demonstrate that direct lineage reprogramming using target cell-type–specific transcription factors can be used to induce NPC-like cells that potentially could be used for autologous cell transplantation-based therapies in the brain or spinal cord.


Nature | 2015

Hallmarks of pluripotency

Alejandro De Los Angeles; Francesco Ferrari; Ruibin Xi; Yuko Fujiwara; Nissim Benvenisty; Hongkui Deng; Rudolf Jaenisch; Soohyun Lee; Harry G. Leitch; M. William Lensch; Ernesto Lujan; Duanqing Pei; Janet Rossant; Marius Wernig; Peter J. Park; George Q. Daley

Stem cells self-renew and generate specialized progeny through differentiation, but vary in the range of cells and tissues they generate, a property called developmental potency. Pluripotent stem cells produce all cells of an organism, while multipotent or unipotent stem cells regenerate only specific lineages or tissues. Defining stem-cell potency relies upon functional assays and diagnostic transcriptional, epigenetic and metabolic states. Here we describe functional and molecular hallmarks of pluripotent stem cells, propose a checklist for their evaluation, and illustrate how forensic genomics can validate their provenance.


Nature | 2015

Early reprogramming regulators identified by prospective isolation and mass cytometry

Ernesto Lujan; Eli R. Zunder; Yi Han Ng; Isabel N. Goronzy; Garry P. Nolan; Marius Wernig

In the context of most induced pluripotent stem (iPS) cell reprogramming methods, heterogeneous populations of non-productive and staggered productive intermediates arise at different reprogramming time points. Despite recent reports claiming substantially increased reprogramming efficiencies using genetically modified donor cells, prospectively isolating distinct reprogramming intermediates remains an important goal to decipher reprogramming mechanisms. Previous attempts to identify surface markers of intermediate cell populations were based on the assumption that, during reprogramming, cells progressively lose donor cell identity and gradually acquire iPS cell properties. Here we report that iPS cell and epithelial markers, such as SSEA1 and EpCAM, respectively, are not predictive of reprogramming during early phases. Instead, in a systematic functional surface marker screen, we find that early reprogramming-prone cells express a unique set of surface markers, including CD73, CD49d and CD200, that are absent in both fibroblasts and iPS cells. Single-cell mass cytometry and prospective isolation show that these distinct intermediates are transient and bridge the gap between donor cell silencing and pluripotency marker acquisition during the early, presumably stochastic, reprogramming phase. Expression profiling reveals early upregulation of the transcriptional regulators Nr0b1 and Etv5 in this reprogramming state, preceding activation of key pluripotency regulators such as Rex1 (also known as Zfp42), Dppa2, Nanog and Sox2. Both factors are required for the generation of the early intermediate state and fully reprogrammed iPS cells, and thus represent some of the earliest known regulators of iPS cell induction. Our study deconvolutes the first steps in a hierarchical series of events that lead to pluripotency acquisition.


Current Opinion in Genetics & Development | 2012

The many roads to Rome: induction of neural precursor cells from fibroblasts

Ernesto Lujan; Marius Wernig

The experimental induction of specific cell fates in related or unrelated lineages has fascinated developmental biologists for decades. The evaluation of altered cell fates in response to ectopic expression during embryonic development has been a standard assay for interrogating gene function. However, until recently examples of cell lineage conversions were limited to closely related and primitive cell types. The induction of pluripotency in fibroblasts prominently highlighted that combinations of transcription factors can be extremely powerful and are much more effective than single genes. On the basis of this conclusion we previously identified transcription factor combinations that directly induce functional neuronal cells from mesodermal and endodermal cells. This work has evoked numerous additional studies demonstrating direct lineage conversion into neural and other lineages. Here, we review the generation of neural progenitor cells from fibroblasts, which is the newest addition to the arena of induced cell types. Surprisingly, two fundamentally different approaches have been taken to induce this cell type, one direct approach and another that involves the intermediate generation of a partially reprogrammed pluripotent state.


Nature | 2015

Failure to replicate the STAP cell phenomenon.

Alejandro De Los Angeles; Francesco Ferrari; Yuko Fujiwara; Ronald Mathieu; Soohyun Lee; Semin Lee; Ho-Chou Tu; Samantha J. Ross; Stephanie S. Chou; Minh Nguyen; Zhaoting Wu; Thorold W. Theunissen; Benjamin E. Powell; Sumeth Imsoonthornruksa; Jiekai Chen; Marti Borkent; Vladislav Krupalnik; Ernesto Lujan; Marius Wernig; Jacob Hanna; Duanqing Pei; Rudolf Jaenisch; Hongkui Deng; Stuart H. Orkin; Peter J. Park; George Q. Daley

Although the reports that stress (such as exposure to acid) can coax somatic cells into a novel state of pluripotency have been retracted, the validity of stimulus-triggered acquisition of pluripotency (STAP) remains unclear (http://dx.doi.org/10.1038/protex. 2014.008 and Supplementary Information). Here we describe the efforts of seven laboratories to replicate STAP, including experiments performed within the laboratory where STAP first originated, as well as re-analysis of the sequencing data from the STAP reports. Neonatal cells treated with two STAP protocols exhibited artefactual autofluoresence rather than bona fide reactivation of an Oct4 (also known as Pou5f1) and green fluorescent protein (GFP) transgene reporter, did not reactivate pluripotency markers towards embryonic stem (ES)-cell-like levels, and failed to generate teratomas or chimaerize blastocysts. Re-analysis of the original RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) data identified discrepancies in the sex and genetic composition of parental donor cells and converted stem cells, and revealed a STAP-derived cell line to be a mixture containing trophoblast stem cells, attesting to the importance of validating the properties and provenance of pluripotent stem cells using a wide range of criteria. To assess the reprogramming capacity of STAP protocols, we used a transgenic Oct4-GFP reporter, which shows GFP reactivation during Oct4/Sox2/Klf4 reprogramming, in established induced pluripotent stem (iPS) cells and in the gonads of mid-gestation ‘all iPS cell’ embryos generated by tetraploid complementation (Extended Data Figs 1 and 2a). Working within the Vacanti laboratory where the concept of STAP cells originated, and assisted by a co-author of the STAP papers, a Daley laboratory member (A.D.L.A.) attempted to replicate two reported STAP protocols: (1) mechanical trituration and acid treatment of mouse lung cells (Brigham and Women’s Hospital (BWH) protocol; see Supplementary Information), and (2) acid treatment of mouse splenocytes (RIKEN protocol; Methods and Extended Data Fig. 2b). Seventy-two hours after stress treatment of lung cells, floating spheres appeared amidst cellular debris. Fluorescence microscopy revealed that both Oct4-GFP and wild-type spheres emitted lowlevel broad spectrum fluorescence detectable within both green and red filters, indicating autofluorescence (Fig. 1a). Untreated Oct4-GFP ES cells did not emit the same low-level broad spectrum fluorescence as STAP-treated cells. STAP-treated splenocytes formed spheres with lower efficiency, but also appeared autofluorescent. Flow cytometry indicated STAP-treated Oct4-GFP cells did not exhibit Oct4-GFP reactivation at levels comparable to control Oct4GFP mouse ES cells, and were indistinguishable from stressed wildtype controls (Fig. 1b). Absence of ES-cell-like levels of Oct4, Sox2 and Nanog transcripts and nonspecific immunofluorescence corroborated flow cytometry data (Extended Data Fig. 2c, d). Rare pluripotent cells should generate teratomas in immunocompromised mice, but STAP cells could not, unlike control ES cells (Extended Data Fig. 2e, f). Replication of the poly-L-glycolic acid (PLGA)-based teratoma production method described in the original STAP reports with GFP cells to distinguish host and donor contribution produced distinct masses of connective tissue, muscle and scar, with minimal GFP content, indicating primarily host origin (Fig. 1c, d and Extended Data Fig. 2g). Rare GFP-positive clusters did not form differentiated tissues characteristic of ES-cell-derived teratomas (Fig. 1d). Autofluorescent spheres failed to enter development after morula aggregation or blastocyst injection (Fig. 1e and Extended Data Fig. 2h–j). Therefore, pluripotency was undetectable in STAP experiments. Six other laboratories (Deng, Hanna, Hochedlinger, Jaenisch, Pei and Wernig) also attempted to generate STAP cells (Table 1) and made the following observations. First, autofluorescent sphere-like aggregates after STAP treatment were universally seen. Second, transgenic reporters used by Obokata and colleagues (GOF18-Oct4-GFP, containing the 18-kilobase genomic Oct4 fragment (GOF18)) and by the Daley, Pei and Hanna laboratories (GOF18-Oct4DPE-GFP, lacking the Oct4 proximal enhancer (PE) element) both exhibit activity in pre-implantation embryos, early post-implantation epiblast cells (embryonic day (E) 5.5), germ cells, and mouse ES/iPS cells; however, differential activity in late post-implantation epiblast (E6.5) and early passage mouse epiblast-derived stem cells has been ascribed to the Oct4 proximal enhancer. Using the same reporter as Obokata and colleagues, the Deng laboratory observed that the GFP signal in chemical iPS cells was easily distinguishable from the autofluorescence of STAP-treated cells (Extended Data Fig. 2k). The Jaenisch, Wernig and Hochedlinger laboratories failed to observe GFP reactivation with Oct4 or Nanog knock-in reporters, excluding a scenario of uncoupling between GFP and endogenous pluripotency expression. Despite a range of tested reporters, no group documented authentic Oct4/Nanog reporter activation that resembled bona fide ES cells. Third, the Deng laboratory failed to observe Oct4, Sox2 and Nanog induction 3 and 7 days after STAP treatment, reducing the likelihood that pluripotency was transiently activated and silenced by day 7 (Extended Data Fig. 2l). Finally, the Hanna, Wernig and Hochedlinger laboratories failed to generate stem-cell lines by culturing STAP-treated cells in leukaemia inhibitory factor (LIF) and adrenocorticotropic hormone (ACTH)-supplemented medium. In summary, 133 replicate attempts failed to document generation of ES-cell-like cells, corroborating and extending a recent report. We re-examined the high-throughput sequencing data from the STAP reports to investigate the genetic provenance of parental CD45 cells and converted STAP cells, STAP stem cells and Fgf4-induced stem cells (FI-SCs) (Fig. 1f). Comparative genomic hybridization array data mentioned in the original paper were not publicly released. Copy number variation (CNV) analysis conducted using ChIP-seq input samples revealed a discrepancy in sex across samples as well as chromosomal aberrations (Fig. 1g). In the original STAP reports, the authors stated that they mixed CD45 cells from male and female mice owing to the small number of CD45 cells retrieved from individual neonatal spleens. However, our analysis indicates that CD45 cells were female, whereas the derived cells (STAP cells, STAP stem cells and FI-SCs) were all male, a clear inconsistency. We note that control ES cells were also male (Fig. 1g). FI-SCs possessed trisomy 8, which renders mouse ES cells germline-incompetent (Fig. 1g). Inferred single nucleotide variants (SNVs) from RNA-seq data allowed classification of samples as genetically similar or dissimilar (Fig. 1h). Control ES cells, parental donor female CD45 cells, STAP cells, and STAP stem cells all possessed similar SNV profiles, consistent with their derivation from a first generation hybrid of C57BL6/129 strains, the reported genotype (Fig. 1h and Extended Data Fig. 3). By contrast, FI-SCs had an SNV profile that matched a single nucleotide polymorphism (SNP) profile of C57BL6 strain origin, indicating


Cell Research | 2010

An imprinted signature helps isolate ESC-equivalent iPSCs.

Ernesto Lujan; Marius Wernig

Institute for Stem Cell Biology and Regenerative Medicine, Department of Pathology; Department of Genetics, Stanford University School of Medicine, 1050 Arastradero Road, Palo Alto, California 94304, USA Cell Research (2010) 20:974-976. doi:10.1038/cr.2010.117; published online 10 August 2010 Cell Research (2010) 20:974-976.


Nature Methods | 2013

An indirect approach to generating specific human cell types

Ernesto Lujan; Marius Wernig

Two groups derived neural and mesodermal cells from human fibroblasts by going through a partially reprogrammed intermediate.


Nature | 2015

Corrigendum: Hallmarks of pluripotency.

Alejandro De Los Angeles; Francesco Ferrari; Ruibin Xi; Yuko Fujiwara; Nissim Benvenisty; Hongkui Deng; Rudolf Jaenisch; Soohyun Lee; Harry G. Leitch; M. William Lensch; Ernesto Lujan; Duanqing Pei; Janet Rossant; Marius Wernig; Peter J. Park; George Q. Daley

This corrects the article DOI: 10.1038/nature15515


Nature | 2015

Corrigendum: Failure to replicate the STAP cell phenomenon.

Alejandro De Los Angeles; Francesco Ferrari; Yuko Fujiwara; Ronald Mathieu; Soohyun Lee; Semin Lee; Ho-Chou Tu; Samantha J. Ross; Stephanie S. Chou; Minh Nguyen; Zhaoting Wu; Thorold W. Theunissen; Benjamin E. Powell; Sumeth Imsoonthornruksa; Jiekai Chen; Marti Borkent; Vladislav Krupalnik; Ernesto Lujan; Marius Wernig; Jacob Hanna; Duanqing Pei; Rudolf Jaenisch; Hongkui Deng; Stuart H. Orkin; Peter J. Park; George Q. Daley

This corrects the article DOI: 10.1038/nature15513


Cell Stem Cell | 2014

m6A RNA Modification Controls Cell Fate Transition in Mammalian Embryonic Stem Cells

Pedro J. Batista; Benoit Molinie; Jinkai Wang; Kun Qu; Jiajing Zhang; Lingjie Li; Donna M. Bouley; Ernesto Lujan; Bahareh Haddad; Kaveh Daneshvar; Ava C. Carter; Ryan A. Flynn; Chan Zhou; Kok-Seong Lim; Peter C. Dedon; Marius Wernig; Alan C. Mullen; Yi Xing; Cosmas Giallourakis; Howard Y. Chang

Collaboration


Dive into the Ernesto Lujan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rudolf Jaenisch

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Duanqing Pei

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge