Network


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

Hotspot


Dive into the research topics where Luis A. Williams is active.

Publication


Featured researches published by Luis A. Williams.


Cell Reports | 2014

Intrinsic Membrane Hyperexcitability of Amyotrophic Lateral Sclerosis Patient-Derived Motor Neurons

Brian J. Wainger; Evangelos Kiskinis; Cassidy Mellin; Ole Wiskow; Steve S.W. Han; Jackson Sandoe; Numa P. Perez; Luis A. Williams; Seungkyu Lee; Gabriella L. Boulting; James D. Berry; Robert H. Brown; Merit Cudkowicz; Bruce P. Bean; Kevin Eggan; Clifford J. Woolf

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease of the motor nervous system. We show using multielectrode array and patch-clamp recordings that hyperexcitability detected by clinical neurophysiological studies of ALS patients is recapitulated in induced pluripotent stem cell-derived motor neurons from ALS patients harboring superoxide dismutase 1 (SOD1), C9orf72, and fused-in-sarcoma mutations. Motor neurons produced from a genetically corrected but otherwise isogenic SOD1(+/+) stem cell line do not display the hyperexcitability phenotype. SOD1(A4V/+) ALS patient-derived motor neurons have reduced delayed-rectifier potassium current amplitudes relative to control-derived motor neurons, a deficit that may underlie their hyperexcitability. The Kv7 channel activator retigabine both blocks the hyperexcitability and improves motor neuron survival in vitro when tested in SOD1 mutant ALS cases. Therefore, electrophysiological characterization of human stem cell-derived neurons can reveal disease-related mechanisms and identify therapeutic candidates.


Cell Stem Cell | 2014

Pathways disrupted in human ALS motor neurons identified through genetic correction of mutant SOD1

Evangelos Kiskinis; Jackson Sandoe; Luis A. Williams; Gabriella L. Boulting; Rob Moccia; Brian J. Wainger; Steve S.W. Han; Theodore Peng; Sebastian Thams; Shravani Mikkilineni; Cassidy Mellin; Florian T. Merkle; Brandi N. Davis-Dusenbery; Michael J. Ziller; Derek Oakley; Justin K. Ichida; Stefania Di Costanzo; Nick Atwater; Morgan L. Maeder; Mathew J. Goodwin; James Nemesh; Robert E. Handsaker; Daniel Paull; Scott Noggle; Steven A. McCarroll; J. Keith Joung; Clifford J. Woolf; Robert H. Brown; Kevin Eggan

Although many distinct mutations in a variety of genes are known to cause Amyotrophic Lateral Sclerosis (ALS), it remains poorly understood how they selectively impact motor neuron biology and whether they converge on common pathways to cause neuronal degeneration. Here, we have combined reprogramming and stem cell differentiation approaches with genome engineering and RNA sequencing to define the transcriptional and functional changes that are induced in human motor neurons by mutant SOD1. Mutant SOD1 protein induced a transcriptional signature indicative of increased oxidative stress, reduced mitochondrial function, altered subcellular transport, and activation of the ER stress and unfolded protein response pathways. Functional studies demonstrated that these pathways were perturbed in a manner dependent on the SOD1 mutation. Finally, interrogation of stem-cell-derived motor neurons produced from ALS patients harboring a repeat expansion in C9orf72 indicates that at least a subset of these changes are more broadly conserved in ALS.


Development | 2014

How to make spinal motor neurons

Brandi N. Davis-Dusenbery; Luis A. Williams; Joseph R. Klim; Kevin Eggan

All muscle movements, including breathing, walking, and fine motor skills rely on the function of the spinal motor neuron to transmit signals from the brain to individual muscle groups. Loss of spinal motor neuron function underlies several neurological disorders for which treatment has been hampered by the inability to obtain sufficient quantities of primary motor neurons to perform mechanistic studies or drug screens. Progress towards overcoming this challenge has been achieved through the synthesis of developmental biology paradigms and advances in stem cell and reprogramming technology, which allow the production of motor neurons in vitro. In this Primer, we discuss how the logic of spinal motor neuron development has been applied to allow generation of motor neurons either from pluripotent stem cells by directed differentiation and transcriptional programming, or from somatic cells by direct lineage conversion. Finally, we discuss methods to evaluate the molecular and functional properties of motor neurons generated through each of these techniques.


Cell | 2012

SnapShot: Directed Differentiation of Pluripotent Stem Cells

Luis A. Williams; Brandi N. Davis-Dusenbery; Kevin Eggan

Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) posses great potential for applications in regenerative medicine, disease modeling, and developmental biology studies. This potential relies on the ability of these cells to differentiate into the hundreds of cell types within the body. Here, we highlight some of strategies for directing the differentiation of ESCs and iPSCs into defined cell types. Most cell types and pathways depicted correspond to published work on human cells, except for the production of spermatozoa, oocyte-like cells, otic hair cells, cortical layers, and optic cup, which were generated with mouse ESCs or iPSCs.In order to uncover these differentiation strategies, stem cell biologists have relied heavily on previous research in model organisms, including


Nature Chemical Biology | 2014

Notch inhibition allows oncogene-independent generation of iPS cells

Justin K. Ichida; Julia Tcw; Luis A. Williams; Ava C. Carter; Yingxiao Shi; Marcelo Tigre Moura; Michael J. Ziller; Sean Singh; Giovanni Amabile; Christoph Bock; Akihiro Umezawa; Lee L. Rubin; James E. Bradner; Hidenori Akutsu; Alexander Meissner; Kevin Eggan

The reprogramming of somatic cells to pluripotency using defined transcription factors holds great promise for biomedicine. However, human reprogramming remains inefficient and relies either on the use of the potentially dangerous oncogenes KLF4 and CMYC or the genetic inhibition of the tumor suppressor gene p53. We hypothesized that inhibition of signal transduction pathways that promote differentiation of the target somatic cells during development might relieve the requirement for non-core pluripotency factors during iPSC reprogramming. Here, we show that inhibition of Notch significantly improves the efficiency of iPSC generation from mouse and human keratinocytes by suppressing p21 in a p53-independent manner and thereby enriching for undifferentiated cells capable of long-term self-renewal. Pharmacological inhibition of Notch enabled routine production of human iPSCs without KLF4 and CMYC while leaving p53 activity intact. Thus, restricting the development of somatic cells by altering intercellular communication enables the production of safer human iPSCs.


Stem cell reports | 2017

Reactive Astrocytes Promote ALS-like Degeneration and Intracellular Protein Aggregation in Human Motor Neurons by Disrupting Autophagy through TGF-β1

Pratibha Tripathi; Natalia Rodriguez-Muela; Joseph R. Klim; A. Sophie de Boer; Sahil Agrawal; Jackson Sandoe; Claudia Lopes; Karolyn Sassi Ogliari; Luis A. Williams; Matthew Shear; Lee L. Rubin; Kevin Eggan; Qiao Zhou

Summary Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly progressing motor neuron disease. Astrocytic factors are known to contribute to motor neuron degeneration and death in ALS. However, the role of astrocyte in promoting motor neuron protein aggregation, a disease hallmark of ALS, remains largely unclear. Here, using culture models of human motor neurons and primary astrocytes of different genotypes (wild-type or SOD1 mutant) and reactive states (non-reactive or reactive), we show that reactive astrocytes, regardless of their genotypes, reduce motor neuron health and lead to moderate neuronal loss. After prolonged co-cultures of up to 2 months, motor neurons show increased axonal and cytoplasmic protein inclusions characteristic of ALS. Reactive astrocytes induce protein aggregation in part by releasing transforming growth factor β1 (TGF-β1), which disrupts motor neuron autophagy through the mTOR pathway. These results reveal the important contribution of reactive astrocytes in promoting aspects of ALS pathology independent of genetic influences.


BMC Plant Biology | 2012

Positive- and negative-acting regulatory elements contribute to the tissue-specific expression of INNER NO OUTER, a YABBY-type transcription factor gene in Arabidopsis

Marissa K. Simon; Luis A. Williams; Kristina Brady-Passerini; Ryan H. Brown; Charles S. Gasser

BackgroundThe INNER NO OUTER (INO) gene, which encodes a YABBY-type transcription factor, specifies and promotes the growth of the outer integument of the ovule in Arabidopsis. INO expression is limited to the abaxial cell layer of the developing outer integument of the ovule and is regulated by multiple regions of the INO promoter, including POS9, a positive element that when present in quadruplicate can produce low-level expression in the normal INO pattern.ResultsSignificant redundancy in activity between different regions of the INO promoter is demonstrated. For specific regulatory elements, multimerization or the addition of the cauliflower mosaic virus 35S general enhancer was able to activate expression of reporter gene constructs that were otherwise incapable of expression on their own. A new promoter element, POS6, is defined and is shown to include sufficient positive regulatory information to reproduce the endogenous pattern of expression in ovules, but other promoter regions are necessary to fully suppress expression outside of ovules. The full-length INO promoter, but not any of the INO promoter deletions tested, is able to act as an enhancer-blocking insulator to prevent the ectopic activation of expression by the 35S enhancer. Sequence conservation between the promoter regions of Arabidopsis thaliana, Brassica oleracea and Brassica rapa aligns closely with the functional definition of the POS6 and POS9 regions, and with a defined INO minimal promoter. The B. oleracea INO promoter is sufficient to promote a similar pattern and level of reporter gene expression in Arabidopsis to that observed for the Arabidopsis promoter.ConclusionsAt least two independent regions of the INO promoter contain sufficient regulatory information to direct the specific pattern but not the level of INO gene expression. These regulatory regions act in a partially redundant manner to promote the expression in a specific pattern in the ovule and suppress expression outside of ovules. Establishment of this pattern requires cooperation and competition between multiple positive and negative regulatory elements.


Scientific Reports | 2018

TDP-43 induces p53-mediated cell death of cortical progenitors and immature neurons

Miriam A. Vogt; Zahra Ehsaei; Philip Knuckles; Adrian Higginbottom; Michaela S. Helmbrecht; Tilo Kunath; Kevin Eggan; Luis A. Williams; Pamela J. Shaw; Wolfgang Wurst; Thomas Floss; Andrea B. Huber; Verdon Taylor

TAR DNA-binding protein 43 (TDP-43) is a key player in neurodegenerative diseases including frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). Accumulation of TDP-43 is associated with neuronal death in the brain. How increased and disease-causing mutant forms of TDP-43 induce cell death remains unclear. Here we addressed the role of TDP-43 during neural development and show that reduced TDP-43 causes defects in neural stem/progenitor cell proliferation but not cell death. However, overexpression of wild type and TDP-43A315T proteins induce p53-dependent apoptosis of neural stem/progenitors and human induced pluripotent cell (iPS)-derived immature cortical neurons. We show that TDP-43 induces expression of the proapoptotic BH3-only genes Bbc3 and Bax, and that p53 inhibition rescues TDP-43 induced cell death of embryonic mouse, and human cortical neurons, including those derived from TDP-43G298S ALS patient iPS cells. Hence, an increase in wild type and mutant TDP-43 induces p53-dependent cell death in neural progenitors developing neurons and this can be rescued. These findings may have important implications for accumulated or mutant TDP-43 induced neurodegenerative diseases.


Molecular Therapy | 2018

A stem cell-based screening platform identifies compounds that desensitize motor neurons to endoplasmic reticulum stress

Sebastian Thams; Emily Rhodes Lowry; Marie-Hélène Larraufie; Krista J. Spiller; Hai Li; Damian J. Williams; Phuong T. Hoang; Elise Jiang; Luis A. Williams; Jackson Sandoe; Kevin Eggan; Ivo Lieberam; Kevin C. Kanning; Brent R. Stockwell; Christopher E. Henderson; Hynek Wichterle

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease selectively targeting motor neurons in the brain and spinal cord. The reasons for differential motor neuron susceptibility remain elusive. We developed a stem cell-based motor neuron assay to study cell-autonomous mechanisms causing motor neuron degeneration, with implications for ALS. A small-molecule screen identified cyclopiazonic acid (CPA) as a stressor to which stem cell-derived motor neurons were more sensitive than interneurons. CPA induced endoplasmic reticulum stress and the unfolded protein response. Furthermore, CPA resulted in an accelerated degeneration of motor neurons expressing human superoxide dismutase 1 (hSOD1) carrying the ALS-causing G93A mutation, compared to motor neurons expressing wild-type hSOD1. A secondary screen identified compounds that alleviated CPA-mediated motor neuron degeneration: three kinase inhibitors and tauroursodeoxycholic acid (TUDCA), a bile acid derivative. The neuroprotective effects of these compounds were validated in human stem cell-derived motor neurons carrying a mutated SOD1 allele (hSOD1A4V). Moreover, we found that the administration of TUDCA in an hSOD1G93A mouse model of ALS reduced muscle denervation. Jointly, these results provide insights into the mechanisms contributing to the preferential susceptibility of ALS motor neurons, and they demonstrate the utility of stem cell-derived motor neurons for the discovery of new neuroprotective compounds.


Neuron | 2014

Axonal Transport of TDP-43 mRNA Granules Is Impaired by ALS-Causing Mutations

Nael H. Alami; Rebecca B. Smith; Monica A. Carrasco; Luis A. Williams; Christina S. Winborn; Steve S.W. Han; Evangelos Kiskinis; Brett J. Winborn; Brian D. Freibaum; Anderson P. Kanagaraj; Alison J. Clare; Nisha M. Badders; Bilada Bilican; Edward Chaum; Siddharthan Chandran; Christopher Shaw; Kevin Eggan; Tom Maniatis; J. Paul Taylor

Collaboration


Dive into the Luis A. Williams'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

Justin K. Ichida

University of Southern California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cassidy Mellin

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Clifford J. Woolf

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge