Network


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

Hotspot


Dive into the research topics where Juan Carlos Izpisua Belmonte is active.

Publication


Featured researches published by Juan Carlos Izpisua Belmonte.


Nature Biotechnology | 2008

Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes

Trond Aasen; Angel Raya; Maria J. Barrero; Elena Garreta; Antonella Consiglio; Federico Gonzalez; Rita Vassena; Josipa Bili cacute; Vladimir Pekarik; Gustavo Tiscornia; Michael J. Edel; Stéphanie Boué; Juan Carlos Izpisua Belmonte

The utility of induced pluripotent stem (iPS) cells for investigating the molecular logic of pluripotency and for eventual clinical application is limited by the low efficiency of current methods for reprogramming. Here we show that reprogramming of juvenile human primary keratinocytes by retroviral transduction with OCT4, SOX2, KLF4 and c-MYC is at least 100-fold more efficient and twofold faster compared with reprogramming of human fibroblasts. Keratinocyte-derived iPS (KiPS) cells appear indistinguishable from human embryonic stem cells in colony morphology, growth properties, expression of pluripotency-associated transcription factors and surface markers, global gene expression profiles and differentiation potential in vitro and in vivo. To underscore the efficiency and practicability of this technology, we generated KiPS cells from single adult human hairs. Our findings provide an experimental model for investigating the bases of cellular reprogramming and highlight potential advantages of using keratinocytes to generate patient-specific iPS cells.


Nature | 2011

SOMATIC CODING MUTATIONS IN HUMAN INDUCED PLURIPOTENT STEM CELLS

Athurva Gore; Zhe Li; Ho Lim Fung; Jessica E. Young; Suneet Agarwal; Jessica Antosiewicz-Bourget; Isabel Canto; Alessandra Giorgetti; Mason A. Israel; Evangelos Kiskinis; Je-Hyuk Lee; Yuin-Han Loh; Philip D. Manos; Nuria Montserrat; Athanasia D. Panopoulos; Sergio Ruiz; Melissa L. Wilbert; Junying Yu; Ewen F. Kirkness; Juan Carlos Izpisua Belmonte; Derrick J. Rossi; James A. Thomson; Kevin Eggan; George Q. Daley; Lawrence S.B. Goldstein; Kun Zhang

Defined transcription factors can induce epigenetic reprogramming of adult mammalian cells into induced pluripotent stem cells. Although DNA factors are integrated during some reprogramming methods, it is unknown whether the genome remains unchanged at the single nucleotide level. Here we show that 22 human induced pluripotent stem (hiPS) cell lines reprogrammed using five different methods each contained an average of five protein-coding point mutations in the regions sampled (an estimated six protein-coding point mutations per exome). The majority of these mutations were non-synonymous, nonsense or splice variants, and were enriched in genes mutated or having causative effects in cancers. At least half of these reprogramming-associated mutations pre-existed in fibroblast progenitors at low frequencies, whereas the rest occurred during or after reprogramming. Thus, hiPS cells acquire genetic modifications in addition to epigenetic modifications. Extensive genetic screening should become a standard procedure to ensure hiPS cell safety before clinical use.


Nature | 2009

Linking the p53 tumour suppressor pathway to somatic cell reprogramming

Teruhisa Kawamura; Jotaro Suzuki; Yunyuan V. Wang; Sergio Menendez; Laura Batlle Morera; Angel Raya; Geoffrey M. Wahl; Juan Carlos Izpisua Belmonte

Reprogramming somatic cells to induced pluripotent stem (iPS) cells has been accomplished by expressing pluripotency factors and oncogenes, but the low frequency and tendency to induce malignant transformation compromise the clinical utility of this powerful approach. We address both issues by investigating the mechanisms limiting reprogramming efficiency in somatic cells. Here we show that reprogramming factors can activate the p53 (also known as Trp53 in mice, TP53 in humans) pathway. Reducing signalling to p53 by expressing a mutated version of one of its negative regulators, by deleting or knocking down p53 or its target gene, p21 (also known as Cdkn1a), or by antagonizing reprogramming-induced apoptosis in mouse fibroblasts increases reprogramming efficiency. Notably, decreasing p53 protein levels enabled fibroblasts to give rise to iPS cells capable of generating germline-transmitting chimaeric mice using only Oct4 (also known as Pou5f1) and Sox2. Furthermore, silencing of p53 significantly increased the reprogramming efficiency of human somatic cells. These results provide insights into reprogramming mechanisms and suggest new routes to more efficient reprogramming while minimizing the use of oncogenes.


Nature | 2010

Zebrafish heart regeneration occurs by cardiomyocyte dedifferentiation and proliferation

Chris Jopling; Eduard Sleep; Marina Raya; Mercè Martí; Angel Raya; Juan Carlos Izpisua Belmonte

Although mammalian hearts show almost no ability to regenerate, there is a growing initiative to determine whether existing cardiomyocytes or progenitor cells can be coaxed into eliciting a regenerative response. In contrast to mammals, several non-mammalian vertebrate species are able to regenerate their hearts, including the zebrafish, which can fully regenerate its heart after amputation of up to 20% of the ventricle. To address directly the source of newly formed cardiomyocytes during zebrafish heart regeneration, we first established a genetic strategy to trace the lineage of cardiomyocytes in the adult fish, on the basis of the Cre/lox system widely used in the mouse. Here we use this system to show that regenerated heart muscle cells are derived from the proliferation of differentiated cardiomyocytes. Furthermore, we show that proliferating cardiomyocytes undergo limited dedifferentiation characterized by the disassembly of their sarcomeric structure, detachment from one another and the expression of regulators of cell-cycle progression. Specifically, we show that the gene product of polo-like kinase 1 (plk1) is an essential component of cardiomyocyte proliferation during heart regeneration. Our data provide the first direct evidence for the source of proliferating cardiomyocytes during zebrafish heart regeneration and indicate that stem or progenitor cells are not significantly involved in this process.


Cell Stem Cell | 2011

Dynamic Changes in the Copy Number of Pluripotency and Cell Proliferation Genes in Human ESCs and iPSCs during Reprogramming and Time in Culture

Louise C. Laurent; Igor Ulitsky; Ileana Slavin; Ha Tran; Andrew J. Schork; Robert Morey; Candace L. Lynch; Julie V. Harness; S.J Lee; Maria J. Barrero; Sherman Ku; Marina Martynova; Ruslan Semechkin; Vasiliy Galat; Joel M. Gottesfeld; Juan Carlos Izpisua Belmonte; Charles E. Murry; Hans S. Keirstead; Hyun Sook Park; Uli Schmidt; Andrew L. Laslett; Franz Josef Müller; Caroline M. Nievergelt; Ron Shamir; Jeanne F. Loring

Genomic stability is critical for the clinical use of human embryonic and induced pluripotent stem cells. We performed high-resolution SNP (single-nucleotide polymorphism) analysis on 186 pluripotent and 119 nonpluripotent samples. We report a higher frequency of subchromosomal copy number variations in pluripotent samples compared to nonpluripotent samples, with variations enriched in specific genomic regions. The distribution of these variations differed between hESCs and hiPSCs, characterized by large numbers of duplications found in a few hESC samples and moderate numbers of deletions distributed across many hiPSC samples. For hiPSCs, the reprogramming process was associated with deletions of tumor-suppressor genes, whereas time in culture was associated with duplications of oncogenic genes. We also observed duplications that arose during a differentiation protocol. Our results illustrate the dynamic nature of genomic abnormalities in pluripotent stem cells and the need for frequent genomic monitoring to assure phenotypic stability and clinical safety.


Nature | 2009

Disease-corrected haematopoietic progenitors from Fanconi anaemia induced pluripotent stem cells

Angel Raya; Ignasi Rodríguez-Pizà; Guillermo Guenechea; Rita Vassena; Susana Navarro; Maria J. Barrero; Antonella Consiglio; Maria Castella; Paula Río; Eduard Sleep; Federico Gonzalez; Gustavo Tiscornia; Elena Garreta; Trond Aasen; Anna Veiga; Inder M. Verma; Jordi Surrallés; Juan A. Bueren; Juan Carlos Izpisua Belmonte

The generation of induced pluripotent stem (iPS) cells has enabled the derivation of patient-specific pluripotent cells and provided valuable experimental platforms to model human disease. Patient-specific iPS cells are also thought to hold great therapeutic potential, although direct evidence for this is still lacking. Here we show that, on correction of the genetic defect, somatic cells from Fanconi anaemia patients can be reprogrammed to pluripotency to generate patient-specific iPS cells. These cell lines appear indistinguishable from human embryonic stem cells and iPS cells from healthy individuals. Most importantly, we show that corrected Fanconi-anaemia-specific iPS cells can give rise to haematopoietic progenitors of the myeloid and erythroid lineages that are phenotypically normal, that is, disease-free. These data offer proof-of-concept that iPS cell technology can be used for the generation of disease-corrected, patient-specific cells with potential value for cell therapy applications.


Developmental Cell | 2001

Dickkopf1 Is Required for Embryonic Head Induction and Limb Morphogenesis in the Mouse

Mahua Mukhopadhyay; Svetlana Shtrom; Concepción Rodríguez-Esteban; Lan Chen; Tohru Tsukui; Lauren Gomer; David W. Dorward; Andrei Glinka; Alexander Grinberg; Sing Ping Huang; Christof Niehrs; Juan Carlos Izpisua Belmonte; Heiner Westphal

Dickkopf1 (Dkk1) is a secreted protein that acts as a Wnt inhibitor and, together with BMP inhibitors, is able to induce the formation of ectopic heads in Xenopus. Here, we show that Dkk1 null mutant embryos lack head structures anterior of the midbrain. Analysis of chimeric embryos implicates the requirement of Dkk1 in anterior axial mesendoderm but not in anterior visceral endoderm for head induction. In addition, mutant embryos show duplications and fusions of limb digits. Characterization of the limb phenotype strongly suggests a role for Dkk1 both in cell proliferation and in programmed cell death. Our data provide direct genetic evidence for the requirement of secreted Wnt antagonists during embryonic patterning and implicate Dkk1 as an essential inducer during anterior specification as well as a regulator during distal limb patterning.


Nature | 2002

Structural basis of BMP signalling inhibition by the cystine knot protein Noggin.

Jay Groppe; Jason Greenwald; Ezra Wiater; Joaquín Rodríguez-León; Aris N. Economides; Witek Kwiatkowski; Markus Affolter; Wylie Vale; Juan Carlos Izpisua Belmonte; Senyon Choe

The interplay between bone morphogenetic proteins (BMPs) and their antagonists governs developmental and cellular processes as diverse as establishment of the embryonic dorsal–ventral axis, induction of neural tissue, formation of joints in the skeletal system and neurogenesis in the adult brain. So far, the three-dimensional structures of BMP antagonists and the structural basis for inactivation have remained unknown. Here we report the crystal structure of the antagonist Noggin bound to BMP-7, which shows that Noggin inhibits BMP signalling by blocking the molecular interfaces of the binding epitopes for both type I and type II receptors. The BMP-7-binding affinity of site-specific variants of Noggin is correlated with alterations in bone formation and apoptosis in chick limb development, showing that Noggin functions by sequestering its ligand in an inactive complex. The scaffold of Noggin contains a cystine (the oxidized form of cysteine) knot topology similar to that of BMPs; thus, ligand and antagonist seem to have evolved from a common ancestral gene.


Nature | 1998

Pitx2 determines left-right asymmetry of internal organs in vertebrates

Aimee K. Ryan; Bruce Blumberg; Concepción Rodríguez-Esteban; Sayuri Yonei-Tamura; Koji Tamura; Tohru Tsukui; Jennifer De La Peña; Walid Sabbagh; Jason Greenwald; Senyon Choe; Dominic P. Norris; Elizabeth J. Robertson; Ronald M. Evans; Michael G. Rosenfeld; Juan Carlos Izpisua Belmonte

The handedness of visceral organs is conserved among vertebrates and is regulated by asymmetric signals relayed by molecules such as Shh, Nodal and activin. The gene Pitx2 is expressed in the left lateral plate mesoderm and, subsequently, in the left heart and gut of mouse, chick and Xenopus embryos. Misexpression of Shh and Nodal induces Pitx2 expression, whereas inhibition of activin signalling blocks it. Misexpression of Pitx2 alters the relative position of organs and the direction of body rotation in chick and Xenopus embryos. Changes in Pitx2 expression are evident in mouse mutants with laterality defects. Thus, Pitx2 seems to serve as a critical downstream transcription target that mediates left–right asymmetry in vertebrates.


Nature Reviews Molecular Cell Biology | 2011

Dedifferentiation, transdifferentiation and reprogramming: three routes to regeneration

Chris Jopling; Stéphanie Boué; Juan Carlos Izpisua Belmonte

The ultimate goal of regenerative medicine is to replace lost or damaged cells. This can potentially be accomplished using the processes of dedifferentiation, transdifferentiation or reprogramming. Recent advances have shown that the addition of a group of genes can not only restore pluripotency in a fully differentiated cell state (reprogramming) but can also induce the cell to proliferate (dedifferentiation) or even switch to another cell type (transdifferentiation). Current research aims to understand how these processes work and to eventually harness them for use in regenerative medicine.

Collaboration


Dive into the Juan Carlos Izpisua Belmonte's collaboration.

Top Co-Authors

Avatar

Guang-Hui Liu

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Concepcion Rodriguez Esteban

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Jun Wu

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Ignacio Sancho-Martinez

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Mo Li

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jing Qu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Fei Yi

Salk Institute for Biological Studies

View shared research outputs
Researchain Logo
Decentralizing Knowledge