Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Cerebellum ; 2022 Oct 03.
Article in English | MEDLINE | ID: mdl-36190676

ABSTRACT

Multiple system atrophy (MSA) is a fatal neurodegenerative disease of unknown etiology characterized by widespread aggregation of the protein alpha-synuclein in neurons and glia. Its orphan status, biological relationship to Parkinson's disease (PD), and rapid progression have sparked interest in drug development. One significant obstacle to therapeutics is disease heterogeneity. Here, we share our process of developing a clinical trial-ready cohort of MSA patients (69 patients in 2 years) within an outpatient clinical setting, and recruiting 20 of these patients into a longitudinal "n-of-few" clinical trial paradigm. First, we deeply phenotype our patients with clinical scales (UMSARS, BARS, MoCA, NMSS, and UPSIT) and tests designed to establish early differential diagnosis (including volumetric MRI, FDG-PET, MIBG scan, polysomnography, genetic testing, autonomic function tests, skin biopsy) or disease activity (PBR06-TSPO). Second, we longitudinally collect biospecimens (blood, CSF, stool) and clinical, biometric, and imaging data to generate antecedent disease-progression scores. Third, in our Mass General Brigham SCiN study (stem cells in neurodegeneration), we generate induced pluripotent stem cell (iPSC) models from our patients, matched to biospecimens, including postmortem brain. We present 38 iPSC lines derived from MSA patients and relevant disease controls (spinocerebellar ataxia and PD, including alpha-synuclein triplication cases), 22 matched to whole-genome sequenced postmortem brain. iPSC models may facilitate matching patients to appropriate therapies, particularly in heterogeneous diseases for which patient-specific biology may elude animal models. We anticipate that deeply phenotyped and genotyped patient cohorts matched to cellular models will increase the likelihood of success in clinical trials for MSA.

3.
Mol Ther ; 27(4): 729-734, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30598301

ABSTRACT

The discovery that ordinary skin cells can be turned into pluripotent stem cells by the forced expression of defined factors has raised hopes that personalized regenerative treatments based on immunologically compatible material derived from a patient's own cells might be realized in the not-too-distant future. A major barrier to the clinical use of induced pluripotent stem cells (iPSCs) was initially presented by the need to employ integrating viral vectors to express the factors that induce an embryonic gene expression profile, which entails potentially oncogenic alteration of the normal genome. Several "non-integrating" reprogramming systems have been developed over the last decade to address this problem. Among these techniques, mRNA reprogramming is the most unambiguously "footprint-free," most productive, and perhaps the best suited to clinical production of stem cells. Herein, we discuss the origins of the mRNA-based reprogramming system, its benefits and drawbacks, recent technical improvements that simplify its application, and the status of current efforts to industrialize this approach to mass-produce human stem cells for the clinic.


Subject(s)
Cellular Reprogramming/genetics , RNA, Messenger/genetics , Cell Differentiation/genetics , Fibroblasts/metabolism , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Nucleosides/genetics , RNA, Small Interfering/genetics , Transfection
4.
Curr Protoc Stem Cell Biol ; 39(1): 4A.6.1-4A.6.25, 2016 Nov.
Article in English | MEDLINE | ID: mdl-31816187

ABSTRACT

Reprogramming somatic cells to generate induced pluripotent stem cells (iPSCs) has presented the biomedical community with a powerful platform to develop new models for human disease. To fully realize the promise of this technology in cell therapy and regenerative medicine, creating iPSCs under current Good Manufacture Practice (cGMP) conditions is paramount. Some reports have described efforts in this regard, resulting in iPSC lines that are cGMP compliant. The technology developed at Allele Biotechnology for footprint-free, feeder-free, and xeno-free reprogramming using only mRNA is very suitable for creating iPSC lines through an established cGMP process. This technology has resulted in a licensing agreement between Allele Biotechnology and Ocata (formerly ACT, now a wholly owned division of Astellas) for clinical applications. All reagents and vessels are certified as cGMP-produced, all equipment and software are certifiable, and all procedures are carried out in Industry ISO 7 or Class 10,000-grade cleanrooms. In this revised version of the unit, we describe the core improvements to implement steps toward cGMP-compliant generation of iPSCs. Recreating a process close to cGMP production in academic research will make these findings more applicable to translational research. © 2016 by John Wiley & Sons, Inc.

5.
Curr Protoc Stem Cell Biol ; 27: 4A.6.1-4A.6.27, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24510287

ABSTRACT

This unit describes a feeder-free protocol for deriving induced pluripotent stem cells (iPSCs) from human fibroblasts by transfection of synthetic mRNA. The reprogramming of somatic cells requires transient expression of a set of transcription factors that collectively activate an endogenous gene regulatory network specifying the pluripotent phenotype. The necessary ectopic factor expression was first effected using retroviruses; however, as viral integration into the genome is problematic for cell therapy applications, the use of footprint-free vectors such as mRNA is increasingly preferred. Strong points of the mRNA approach include high efficiency, rapid kinetics, and obviation of a clean-up phase to purge the vector. Still, the method is relatively laborious and has, up to now, involved the use of feeder cells, which brings drawbacks including poor applicability to clinically oriented iPSC derivation. Using the methods described here, mRNA reprogramming can be performed without feeders at much-reduced labor and material costs relative to established protocols.


Subject(s)
Cellular Reprogramming , Feeder Cells , Fibroblasts/cytology , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , RNA, Messenger/metabolism , Amino Acid Sequence , Base Sequence , Colony-Forming Units Assay , Humans , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Plasmids/metabolism , Staining and Labeling , Templates, Genetic , Transcription, Genetic
6.
Sci Rep ; 2: 657, 2012.
Article in English | MEDLINE | ID: mdl-22984641

ABSTRACT

The therapeutic promise of induced pluripotent stem cells (iPSCs) has spurred efforts to circumvent genome alteration when reprogramming somatic cells to pluripotency. Approaches based on episomal DNA, Sendai virus, and messenger RNA (mRNA) can generate "footprint-free" iPSCs with efficiencies equaling or surpassing those attained with integrating viral vectors. The mRNA method uniquely affords unprecedented control over reprogramming factor (RF) expression while obviating a cleanup phase to purge residual traces of vector. Currently, mRNA-based reprogramming is relatively laborious due to the need to transfect daily for ~2 weeks to induce pluripotency, and requires the use of feeder cells that add complexity and variability to the procedure while introducing a route for contamination with non-human-derived biological material. We accelerated the mRNA reprogramming process through stepwise optimization of the RF cocktail and leveraged these kinetic gains to establish a feeder-free, xeno-free protocol which slashes the time, cost and effort involved in iPSC derivation.


Subject(s)
Induced Pluripotent Stem Cells/physiology , RNA, Messenger/physiology , Cell Proliferation , Cell Shape , Cell Survival , Cells, Cultured , Coculture Techniques , Herpes Simplex Virus Protein Vmw65/genetics , Homeodomain Proteins/genetics , Humans , MyoD Protein/genetics , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics
7.
Cell Stem Cell ; 7(5): 618-30, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20888316

ABSTRACT

Clinical application of induced pluripotent stem cells (iPSCs) is limited by the low efficiency of iPSC derivation and the fact that most protocols modify the genome to effect cellular reprogramming. Moreover, safe and effective means of directing the fate of patient-specific iPSCs toward clinically useful cell types are lacking. Here we describe a simple, nonintegrating strategy for reprogramming cell fate based on administration of synthetic mRNA modified to overcome innate antiviral responses. We show that this approach can reprogram multiple human cell types to pluripotency with efficiencies that greatly surpass established protocols. We further show that the same technology can be used to efficiently direct the differentiation of RNA-induced pluripotent stem cells (RiPSCs) into terminally differentiated myogenic cells. This technology represents a safe, efficient strategy for somatic cell reprogramming and directing cell fate that has broad applicability for basic research, disease modeling, and regenerative medicine.


Subject(s)
Cell Differentiation/drug effects , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , RNA, Messenger , Cell Lineage , Cells, Cultured , Humans , RNA, Messenger/pharmacology
8.
Mech Ageing Dev ; 130(1-2): 46-53, 2009.
Article in English | MEDLINE | ID: mdl-18479735

ABSTRACT

The effector cells of the blood have limited lifetimes and must be replenished continuously throughout life from a small reserve of hematopoietic stem cells (HSCs) in the bone marrow. Although serial bone marrow transplantation experiments in mice suggest that the replicative potential of HSCs is finite, there is little evidence that replicative senescence causes depletion of the stem cell pool during the normal lifespan of either mouse or man. Studies conducted in murine genetic models defective in DNA repair, intracellular ROS management, and telomere maintenance indicate that all these pathways are critical to the longevity and stress response of the stem cell pool. With age, HSCs show an increased propensity to differentiate towards myeloid rather than lymphoid lineages, which may contribute to the decline in lymphopoiesis that attends aging. Challenges for the future include assessing the significance of 'lineage skewing' to immune dysfunction, and investigating the role of epigenetic dysregulation in HSC aging.


Subject(s)
Aging/genetics , Genomics , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Aged , Aging/physiology , Animals , Hematopoiesis/physiology , Humans
9.
Aging Cell ; 6(6): 775-82, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17925006

ABSTRACT

It has been proposed that cumulative somatic mutations contribute to the aging process by disrupting the transcriptional networks that regulate cell structure and function. Experimental support for this model emerged from a recent study of cardiomyocytes that showed a dramatic increase in the transcriptional heterogeneity of these long-lived postmitotic cells with age. To determine if regulatory instability is a hallmark of aging in renewing tissues, we evaluated gene expression noise in four hematopoietic cell types: stem cells, granulocytes, naïve B cells and naïve T cells. We used flow cytometry to purify phenotypically equivalent cells from young and old mice, and applied multiplexed quantitative reverse transcription-polymerase chain reaction to measure the copy number of six different mRNA transcripts in 324 individual cells. There was a trend toward higher transcript levels in cells isolated from old animals, but no significant increase in transcriptional heterogeneity with age was found in the surveyed populations. Flow cytometric analysis of membrane protein expression also indicated that cell-to-cell variability was unaffected by age. We conclude that large-scale regulatory destabilization is not a universal concomitant of aging, and may be of significance as an aging mechanism primarily in nonrenewing tissues.


Subject(s)
Aging/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Animals , Cellular Senescence/genetics , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis
10.
Proc Natl Acad Sci U S A ; 103(47): 17807-12, 2006 Nov 21.
Article in English | MEDLINE | ID: mdl-17098862

ABSTRACT

We report here a systematic, quantitative population analysis of transcription factor expression within developmental progenitors, made possible by a microfluidic chip-based "digital RT-PCR" assay that can count template molecules in cDNA samples prepared from single cells. In a survey encompassing five classes of early hematopoietic precursor, we found markedly heterogeneous expression of the transcription factor PU.1 in hematopoietic stem cells and divergent patterns of PU.1 expression within flk2- and flk2+ common myeloid progenitors. The survey also revealed significant differences in the level of the housekeeping transcript GAPDH across the surveyed populations, which demonstrates caveats of normalizing expression data to endogenous controls and underscores the need to put gene measurement on an absolute, copy-per-cell basis.


Subject(s)
Gene Expression Profiling , Hematopoietic Stem Cells/physiology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/metabolism , Animals , Gene Expression Profiling/instrumentation , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis/instrumentation , Oligonucleotide Array Sequence Analysis/methods , Transcription Factors/genetics
11.
Curr Opin Immunol ; 15(2): 166-75, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12633666

ABSTRACT

During lymphopoiesis, precursor cells negotiate a complex regulatory space, defined by the levels of several competing and cross-regulating transcription factors, before arriving at stable states of commitment to the B-, T- and NK-specific developmental programs. Recent perturbation experiments provide evidence that this space has three major axes, corresponding to the PU.1 versus GATA-1 balance, the intensity of Notch signaling through the CSL pathway, and the ratio of E-box transcription factors to their Id protein antagonists.


Subject(s)
Cell Lineage/physiology , Lymphocytes/physiology , Transcription Factors/physiology , Animals , Hematopoiesis/immunology , Hematopoiesis/physiology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/physiology , Humans , Lymphocytes/immunology , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...