Network


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

Hotspot


Dive into the research topics where W. Travis Berggren is active.

Publication


Featured researches published by W. Travis Berggren.


Nature Biotechnology | 2006

Derivation of human embryonic stem cells in defined conditions

Tenneille E. Ludwig; Mark E. Levenstein; Jeffrey M. Jones; W. Travis Berggren; Erika R Mitchen; Jennifer L. Frane; Leann J Crandall; Christine A. Daigh; Kevin R. Conard; Marian S Piekarczyk; Rachel A. Llanas; James A. Thomson

We have previously reported that high concentrations of basic fibroblast growth factor (bFGF) support feeder-independent growth of human embryonic stem (ES) cells, but those conditions included poorly defined serum and matrix components. Here we report feeder-independent human ES cell culture that includes protein components solely derived from recombinant sources or purified from human material. We describe the derivation of two new human ES cell lines in these defined culture conditions.


Cell Research | 2012

The metabolome of induced pluripotent stem cells reveals metabolic changes occurring in somatic cell reprogramming

Athanasia D. Panopoulos; Oscar Yanes; Sergio Ruiz; Yasuyuki S. Kida; Dinh Diep; Ralf Tautenhahn; Aída Herrerías; Erika M. Batchelder; Nongluk Plongthongkum; Margaret Lutz; W. Travis Berggren; Kun Zhang; Ronald M. Evans; Gary Siuzdak; Juan Carlos Izpisua Belmonte

Metabolism is vital to every aspect of cell function, yet the metabolome of induced pluripotent stem cells (iPSCs) remains largely unexplored. Here we report, using an untargeted metabolomics approach, that human iPSCs share a pluripotent metabolomic signature with embryonic stem cells (ESCs) that is distinct from their parental cells, and that is characterized by changes in metabolites involved in cellular respiration. Examination of cellular bioenergetics corroborated with our metabolomic analysis, and demonstrated that somatic cells convert from an oxidative state to a glycolytic state in pluripotency. Interestingly, the bioenergetics of various somatic cells correlated with their reprogramming efficiencies. We further identified metabolites that differ between iPSCs and ESCs, which revealed novel metabolic pathways that play a critical role in regulating somatic cell reprogramming. Our findings are the first to globally analyze the metabolome of iPSCs, and provide mechanistic insight into a new layer of regulation involved in inducing pluripotency, and in evaluating iPSC and ESC equivalence.


Nature | 2016

In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration

Keiichiro Suzuki; Yuji Tsunekawa; Reyna Hernández-Benítez; Jun Wu; Jie Zhu; Euiseok J. Kim; Fumiyuki Hatanaka; Mako Yamamoto; Toshikazu Araoka; Zhe Li; Masakazu Kurita; Tomoaki Hishida; Mo Li; Emi Aizawa; Shicheng Guo; Song Chen; April Goebl; Rupa Devi Soligalla; Jing Qu; Tingshuai Jiang; Xin Fu; Maryam Jafari; Concepcion Rodriguez Esteban; W. Travis Berggren; Jeronimo Lajara; Estrella Núñez-Delicado; Pedro Guillen; Josep M. Campistol; Fumio Matsuzaki; Guang-Hui Liu

Targeted genome editing via engineered nucleases is an exciting area of biomedical research and holds potential for clinical applications. Despite rapid advances in the field, in vivo targeted transgene integration is still infeasible because current tools are inefficient, especially for non-dividing cells, which compose most adult tissues. This poses a barrier for uncovering fundamental biological principles and developing treatments for a broad range of genetic disorders. Based on clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) technology, here we devise a homology-independent targeted integration (HITI) strategy, which allows for robust DNA knock-in in both dividing and non-dividing cells in vitro and, more importantly, in vivo (for example, in neurons of postnatal mammals). As a proof of concept of its therapeutic potential, we demonstrate the efficacy of HITI in improving visual function using a rat model of the retinal degeneration condition retinitis pigmentosa. The HITI method presented here establishes new avenues for basic research and targeted gene therapies.


Current Biology | 2011

A high proliferation rate is required for cell reprogramming and maintenance of human embryonic stem cell identity.

Sergio Ruiz; Athanasia D. Panopoulos; Aída Herrerías; Karl-Dimiter Bissig; Margaret Lutz; W. Travis Berggren; Inder M. Verma; Juan Carlos Izpisua Belmonte

Human embryonic stem (hES) cells show an atypical cell-cycle regulation characterized by a high proliferation rate and a short G1 phase. In fact, a shortened G1 phase might protect ES cells from external signals inducing differentiation, as shown for certain stem cells. It has been suggested that self-renewal and pluripotency are intimately linked to cell-cycle regulation in ES cells, although little is known about the overall importance of the cell-cycle machinery in maintaining ES cell identity. An appealing model to address whether the acquisition of stem cell properties is linked to cell-cycle regulation emerged with the ability to generate induced pluripotent stem (iPS) cells by expression of defined transcription factors. Here, we show that the characteristic cell-cycle signature of hES cells is acquired as an early event in cell reprogramming. We demonstrate that induction of cell proliferation increases reprogramming efficiency, whereas cell-cycle arrest inhibits successful reprogramming. Furthermore, we show that cell-cycle arrest is sufficient to drive hES cells toward irreversible differentiation. Our results establish a link that intertwines the mechanisms of cell-cycle control with the mechanisms underlying the acquisition and maintenance of ES cell identity.


Nature | 2012

Increased proteasome activity in human embryonic stem cells is regulated by PSMD11

David Vilchez; Leah Boyer; Ianessa Morantte; Margaret Lutz; Carsten Merkwirth; Derek Joyce; Brian Spencer; Lesley J. Page; Eliezer Masliah; W. Travis Berggren; Fred H. Gage; Andrew Dillin

Embryonic stem cells can replicate continuously in the absence of senescence and, therefore, are immortal in culture. Although genome stability is essential for the survival of stem cells, proteome stability may have an equally important role in stem-cell identity and function. Furthermore, with the asymmetric divisions invoked by stem cells, the passage of damaged proteins to daughter cells could potentially destroy the resulting lineage of cells. Therefore, a firm understanding of how stem cells maintain their proteome is of central importance. Here we show that human embryonic stem cells (hESCs) exhibit high proteasome activity that is correlated with increased levels of the 19S proteasome subunit PSMD11 (known as RPN-6 in Caenorhabditis elegans) and a corresponding increased assembly of the 26S/30S proteasome. Ectopic expression of PSMD11 is sufficient to increase proteasome assembly and activity. FOXO4, an insulin/insulin-like growth factor-I (IGF-I) responsive transcription factor associated with long lifespan in invertebrates, regulates proteasome activity by modulating the expression of PSMD11 in hESCs. Proteasome inhibition in hESCs affects the expression of pluripotency markers and the levels of specific markers of the distinct germ layers. Our results suggest a new regulation of proteostasis in hESCs that links longevity and stress resistance in invertebrates to hESC function and identity.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Human and mouse adipose-derived cells support feeder-independent induction of pluripotent stem cells

Shigeki Sugii; Yasuyuki S. Kida; Teruhisa Kawamura; Jotaro Suzuki; Rita Vassena; Yun-Qiang Yin; Margaret Lutz; W. Travis Berggren; Juan Carlos Izpisúa Belmonte; Ronald M. Evans

Although adipose tissue is an expandable and readily attainable source of proliferating, multipotent stem cells, its potential for use in regenerative medicine has not been extensively explored. Here we report that adult human and mouse adipose-derived stem cells can be reprogrammed to induced pluripotent stem (iPS) cells with substantially higher efficiencies than those reported for human and mouse fibroblasts. Unexpectedly, both human and mouse iPS cells can be obtained in feeder-free conditions. We discovered that adipose-derived stem cells intrinsically express high levels of pluripotency factors such as basic FGF, TGFβ, fibronectin, and vitronectin and can serve as feeders for both autologous and heterologous pluripotent cells. These results demonstrate a great potential for adipose-derived cells in regenerative therapeutics and as a model for studying the molecular mechanisms of feeder-free iPS generation and maintenance.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Mass spectrometry identifies and quantifies 74 unique histone H4 isoforms in differentiating human embryonic stem cells

Doug Phanstiel; Justin Brumbaugh; W. Travis Berggren; Kevin R. Conard; Xuezhu Feng; Mark E. Levenstein; Graeme C. McAlister; James A. Thomson; Joshua J. Coon

Epigenetic regulation through chromatin is thought to play a critical role in the establishment and maintenance of pluripotency. Traditionally, antibody-based technologies were used to probe for specific posttranslational modifications (PTMs) present on histone tails, but these methods do not generally reveal the presence of multiple modifications on a single-histone tail (combinatorial codes). Here, we describe technology for the discovery and quantification of histone combinatorial codes that is based on chromatography and mass spectrometry. We applied this methodology to decipher 74 discrete combinatorial codes on the tail of histone H4 from human embryonic stem (ES) cells. Finally, we quantified the abundances of these codes as human ES cells undergo differentiation to reveal striking changes in methylation and acetylation patterns. For example, H4R3 methylation was observed only in the presence of H4K20 dimethylation; such context-specific patterning exemplifies the power of this technique.


Journal of Proteome Research | 2008

A Proteomics Grade Electron Transfer Dissociation-enabled Hybrid Linear Ion Trap-orbitrap Mass Spectrometer

Graeme C. McAlister; W. Travis Berggren; Jens Griep-Raming; Stevan Horning; Alexander Makarov; Doug Phanstiel; George C. Stafford; Danielle L. Swaney; John Edward Philip Syka; Joshua J. Coon

Here we detail the modification of a quadrupole linear ion trap-orbitrap hybrid (QLT-orbitrap) mass spectrometer to accommodate a negative chemical ionization (NCI) source. The NCI source is used to produce fluoranthene radical anions for imparting electron transfer dissociation (ETD). The anion beam is stable, robust, and intense so that a sufficient amount of reagents can be injected into the QLT in only 4-8 ms. Following ion/ion reaction in the QLT, ETD product ions are mass-to-charge (m/z) analyzed in either the QLT (for speed and sensitivity) or the orbitrap (for mass resolution and accuracy). Here we describe the physical layout of this device, parametric optimization of anion transport, an evaluation of relevant ETD figures of merit, and the application of this instrument to protein sequence analysis. Described proteomic applications include complex peptide mixture analysis, post-translational modification (PTM) site identification, isotope-encoded quantitation, large peptide characterization, and intact protein analysis. From these experiments, we conclude the ETD-enabled orbitrap will provide the proteomic field with several new opportunities and represents an advance in protein sequence analysis technologies.


Cell | 2017

Interspecies Chimerism with Mammalian Pluripotent Stem Cells

Jun Wu; Aida Platero-Luengo; Masahiro Sakurai; Atsushi Sugawara; M.A. Gil; Takayoshi Yamauchi; Keiichiro Suzuki; Y. S. Bogliotti; C. Cuello; Mariana Morales Valencia; Daiji Okumura; Jingping Luo; Marcela Vilarino; I. Parrilla; Delia Alba Soto; Cristina A. Martinez; Tomoaki Hishida; Sonia Sánchez-Bautista; M. Llanos Martinez-Martinez; Huili Wang; A. Nohalez; Emi Aizawa; Paloma Martínez-Redondo; Alejandro Ocampo; Pradeep Reddy; Jordi Roca; Elizabeth A. Maga; Concepcion Rodriguez Esteban; W. Travis Berggren; Estrella Nuñez Delicado

Interspecies blastocyst complementation enables organ-specific enrichment of xenogenic pluripotent stem cell (PSC) derivatives. Here, we establish a versatile blastocyst complementation platform based on CRISPR-Cas9-mediated zygote genome editing and show enrichment of rat PSC-derivatives in several tissues of gene-edited organogenesis-disabled mice. Besides gaining insights into species evolution, embryogenesis, and human disease, interspecies blastocyst complementation might allow human organ generation in animals whose organ size, anatomy, and physiology are closer to humans. To date, however, whether human PSCs (hPSCs) can contribute to chimera formation in non-rodent species remains unknown. We systematically evaluate the chimeric competency of several types of hPSCs using a more diversified clade of mammals, the ungulates. We find that naïve hPSCs robustly engraft in both pig and cattle pre-implantation blastocysts but show limited contribution to post-implantation pig embryos. Instead, an intermediate hPSC type exhibits higher degree of chimerism and is able to generate differentiated progenies in post-implantation pig embryos.


Stem Cells | 2011

Brief Report: Efficient Generation of Hematopoietic Precursors and Progenitors from Human Pluripotent Stem Cell Lines

Niels-Bjarne Woods; Aaron S. Parker; Roksana Moraghebi; Margaret Lutz; Amy L. Firth; Kristen J. Brennand; W. Travis Berggren; Angel Raya; Juan Carlos Izpisua Belmonte; Fred H. Gage; Inder M. Verma

By mimicking embryonic development of the hematopoietic system, we have developed an optimized in vitro differentiation protocol for the generation of precursors of hematopoietic lineages and primitive hematopoietic cells from human embryonic stem cells (ESC) and induced pluripotent stem cells (iPSCs). Factors such as cytokines, extra cellular matrix components, and small molecules as well as the temporal association and concentration of these factors were tested on seven different human ESC and iPSC lines. We report the differentiation of up to 84% human CD45+ cells (average 41% ± 16%, from seven pluripotent lines) from the differentiation culture, including significant numbers of primitive CD45+/CD34+ and CD45+/CD34+/CD38− hematopoietic progenitors. Moreover, the numbers of hematopoietic progenitor cells generated, as measured by colony forming unit assays, were comparable to numbers obtained from fresh umbilical cord blood mononuclear cell isolates on a per CD45+ cell basis. Our approach demonstrates highly efficient generation of multipotent hematopoietic progenitors with among the highest efficiencies reported to date (CD45+/CD34+) using a single standardized differentiation protocol on several human ESC and iPSC lines. Our data add to the cumulating evidence for the existence of an in vitro derived precursor to the hematopoietic stem cell (HSC) with limited engrafting ability in transplanted mice but with multipotent hematopoietic potential. Because this protocol efficiently expands the preblood precursors and hematopoietic progenitors, it is ideal for testing novel factors for the generation and expansion of definitive HSCs with long‐term repopulating ability. STEM CELLS 2011;29:1158–1164

Collaboration


Dive into the W. Travis Berggren's collaboration.

Top Co-Authors

Avatar

Lloyd M. Smith

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Margaret Lutz

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Athanasia D. Panopoulos

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Juan Carlos Izpisua Belmonte

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Doug Phanstiel

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Graeme C. McAlister

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Joshua J. Coon

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Ronald M. Evans

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Veronica Modesto

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge