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










Publication year range
1.
Cells ; 13(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38891104

ABSTRACT

Mutations in the DMD gene cause fatal Duchenne Muscular Dystrophy (DMD). An attractive therapeutic approach is autologous cell transplantation utilizing myogenic progenitors derived from induced pluripotent stem cells (iPSCs). Given that a significant number of DMD mutations occur between exons 45 and 55, we developed a gene knock-in approach to correct any mutations downstream of exon 44. We applied this approach to two DMD patient-specific iPSC lines carrying mutations in exons 45 and 51 and confirmed mini-DYSTROPHIN (mini-DYS) protein expression in corrected myotubes by western blot and immunofluorescence staining. Transplantation of gene-edited DMD iPSC-derived myogenic progenitors into NSG/mdx4Cv mice produced donor-derived myofibers, as shown by the dual expression of human DYSTROPHIN and LAMIN A/C. These findings further provide proof-of-concept for the use of programmable nucleases for the development of autologous iPSC-based therapy for muscular dystrophies.


Subject(s)
Dystrophin , Gene Editing , Induced Pluripotent Stem Cells , Muscular Dystrophy, Duchenne , Mutation , Animals , Humans , Mice , CRISPR-Cas Systems/genetics , Dystrophin/genetics , Dystrophin/metabolism , Exons/genetics , Gene Editing/methods , Induced Pluripotent Stem Cells/metabolism , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Muscular Dystrophy, Duchenne/pathology
2.
NPJ Regen Med ; 9(1): 16, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38575647

ABSTRACT

Pluripotent stem cell (PSC)-based cell therapy is an attractive option for the treatment of multiple human disorders, including muscular dystrophies. While in vitro differentiating PSCs can generate large numbers of human lineage-specific tissue, multiple studies evidenced that these cell populations mostly display embryonic/fetal features. We previously demonstrated that transplantation of PSC-derived myogenic progenitors provides long-term engraftment and functional improvement in several dystrophic mouse models, but it remained unknown whether donor-derived myofibers mature to match adult tissue. Here, we transplanted iPAX7 myogenic progenitors into muscles of non-dystrophic and dystrophic mice and compared the transcriptional landscape of human grafts with respective in vitro-differentiated iPAX7 myotubes as well as human skeletal muscle biospecimens. Pairing bulk RNA sequencing with computational deconvolution of human reads, we were able to pinpoint key myogenic changes that occur during the in vitro-to-in vivo transition, confirm developmental maturity, and consequently evaluate their applicability for cell-based therapies.

3.
Cells ; 12(8)2023 04 13.
Article in English | MEDLINE | ID: mdl-37190056

ABSTRACT

Pluripotent stem (PS) cells enable the scalable production of tissue-specific derivatives with therapeutic potential for various clinical applications, including muscular dystrophies. Given the similarity to human counterparts, the non-human primate (NHP) is an ideal preclinical model to evaluate several questions, including delivery, biodistribution, and immune response. While the generation of human-induced PS (iPS)-cell-derived myogenic progenitors is well established, there have been no data for NHP counterparts, probably due to the lack of an efficient system to differentiate NHP iPS cells towards the skeletal muscle lineage. Here, we report the generation of three independent Macaca fascicularis iPS cell lines and their myogenic differentiation using PAX7 conditional expression. The whole-transcriptome analysis confirmed the successful sequential induction of mesoderm, paraxial mesoderm, and myogenic lineages. NHP myogenic progenitors efficiently gave rise to myotubes under appropriate in vitro differentiation conditions and engrafted in vivo into the TA muscles of NSG and FKRP-NSG mice. Lastly, we explored the preclinical potential of these NHP myogenic progenitors in a single wild-type NHP recipient, demonstrating engraftment and characterizing the interaction with the host immune response. These studies establish an NHP model system through which iPS-cell-derived myogenic progenitors can be studied.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Animals , Mice , Induced Pluripotent Stem Cells/metabolism , Tissue Distribution , Pluripotent Stem Cells/metabolism , Muscle, Skeletal/metabolism , Primates , Pentosyltransferases/metabolism
5.
Nat Cell Biol ; 24(4): 538-553, 2022 04.
Article in English | MEDLINE | ID: mdl-35411081

ABSTRACT

Skeletal muscle has long been recognized as an inhospitable site for disseminated tumour cells (DTCs). Yet its antimetastatic nature has eluded a thorough mechanistic examination. Here, we show that DTCs traffic to and persist within skeletal muscle in mice and in humans, which raises the question of how this tissue suppresses colonization. Results from mouse and organotypic culture models along with metabolomic profiling suggested that skeletal muscle imposes a sustained oxidative stress on DTCs that impairs their proliferation. Functional studies demonstrated that disrupting reduction-oxidation homeostasis via chemogenetic induction of reactive oxygen species slowed proliferation in a more fertile organ: the lung. Conversely, enhancement of the antioxidant potential of tumour cells through ectopic expression of catalase in the tumour or host mitochondria allowed robust colonization of skeletal muscle. These findings reveal a profound metabolic bottleneck imposed on DTCs and sustained by skeletal muscle. A thorough understanding of this biology could reveal previously undocumented DTC vulnerabilities that can be exploited to prevent metastasis in other more susceptible tissues.


Subject(s)
Neoplasms , Oxidative Stress , Animals , Mice , Muscle, Skeletal/metabolism , Neoplasms/metabolism , Oxidation-Reduction , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism
6.
Annu Rev Pathol ; 16: 409-432, 2021 01 24.
Article in English | MEDLINE | ID: mdl-33276706

ABSTRACT

Over the last four decades, the cancer biology field has concentrated on cellular and microenvironmental drivers of metastasis. Despite this focus, mortality rates upon diagnosis of metastatic disease remain essentially unchanged. Would a small change in perspective help? Knowing what constitutes an inhospitable, rather than hospitable, microenvironment could provide the inspiration necessary to develop better therapies and preventative strategies. In this review, we canvas the literature for hints about what characteristics four common antimetastatic niches-skeletal muscle, spleen, thyroid, and yellow bone marrow-have in common. We posit that thorough molecular and mechanistic characterization of antimetastatic tissues may inspire reimagined therapies that inhibit metastatic development and/or progression in an enduring manner.


Subject(s)
Neoplasm Invasiveness/pathology , Neoplasms/pathology , Tumor Microenvironment , Animals , Humans
7.
Nat Cell Biol ; 21(2): 238-250, 2019 02.
Article in English | MEDLINE | ID: mdl-30664790

ABSTRACT

The presence of disseminated tumour cells (DTCs) in bone marrow is predictive of poor metastasis-free survival of patients with breast cancer with localized disease. DTCs persist in distant tissues despite systemic administration of adjuvant chemotherapy. Many assume that this is because the majority of DTCs are quiescent. Here, we challenge this notion and provide evidence that the microenvironment of DTCs protects them from chemotherapy, independent of cell cycle status. We show that chemoresistant DTCs occupy the perivascular niche (PVN) of distant tissues, where they are protected from therapy by vascular endothelium. Inhibiting integrin-mediated interactions between DTCs and the PVN, driven partly by endothelial-derived von Willebrand factor and vascular cell adhesion molecule 1, sensitizes DTCs to chemotherapy. Importantly, chemosensitization is achieved without inducing DTC proliferation or exacerbating chemotherapy-associated toxicities, and ultimately results in prevention of bone metastasis. This suggests that prefacing adjuvant therapy with integrin inhibitors is a viable clinical strategy to eradicate DTCs and prevent metastasis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Blood Vessels/drug effects , Mammary Neoplasms, Experimental/drug therapy , Tumor Microenvironment/drug effects , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Female , Integrins/metabolism , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Transgenic , Paclitaxel/administration & dosage
8.
Cell Syst ; 6(3): 268-270, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29596781

ABSTRACT

Watson et al. use microenvironment microarrays to assess how extrinsic signals within the tumor microenvironment influence HER2++ breast cancer resistance to the HER2-targeted tyrosine kinase inhibitor lapatinib.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Cell Line, Tumor , Friends , Humans , Quinazolines , Receptor, ErbB-2 , Tumor Microenvironment
9.
Elife ; 62017 05 05.
Article in English | MEDLINE | ID: mdl-28475037

ABSTRACT

NEDD8 is a ubiquitin-like modifier most well-studied for its role in activating the largest family of ubiquitin E3 ligases, the cullin-RING ligases (CRLs). While many non-cullin neddylation substrates have been proposed over the years, validation of true NEDD8 targets has been challenging, as overexpression of exogenous NEDD8 can trigger NEDD8 conjugation through the ubiquitylation machinery. Here, we developed a deconjugation-resistant form of NEDD8 to stabilize the neddylated form of cullins and other non-cullin substrates. Using this strategy, we identified Ubc12, a NEDD8-specific E2 conjugating enzyme, as a substrate for auto-neddylation. Furthermore, we characterized SENP8/DEN1 as the protease that counteracts Ubc12 auto-neddylation, and observed aberrant neddylation of Ubc12 and other NEDD8 conjugation pathway components in SENP8-deficient cells. Importantly, loss of SENP8 function contributes to accumulation of CRL substrates and defective cell cycle progression. Thus, our study highlights the importance of SENP8 in maintaining proper neddylation levels for CRL-dependent proteostasis.


Subject(s)
Endopeptidases/metabolism , NEDD8 Protein/metabolism , Protein Processing, Post-Translational , Ubiquitin-Conjugating Enzymes/metabolism , Cell Cycle , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans
10.
Mol Cell ; 58(2): 323-38, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25843623

ABSTRACT

Excess dormant origins bound by the minichromosome maintenance (MCM) replicative helicase complex play a critical role in preventing replication stress, chromosome instability, and tumorigenesis. In response to DNA damage, replicating cells must coordinate DNA repair and dormant origin firing to ensure complete and timely replication of the genome; how cells regulate this process remains elusive. Herein, we identify a member of the Fanconi anemia (FA) DNA repair pathway, FANCI, as a key effector of dormant origin firing in response to replication stress. Cells lacking FANCI have reduced number of origins, increased inter-origin distances, and slowed proliferation rates. Intriguingly, ATR-mediated FANCI phosphorylation inhibits dormant origin firing while promoting replication fork restart/DNA repair. Using super-resolution microscopy, we show that FANCI co-localizes with MCM-bound chromatin in response to replication stress. These data reveal a unique role for FANCI as a modulator of dormant origin firing and link timely genome replication to DNA repair.


Subject(s)
Chromatin/metabolism , DNA Damage , DNA Replication , Fanconi Anemia Complementation Group Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Proliferation , Fanconi Anemia Complementation Group Proteins/genetics , HeLa Cells , Humans , Hydroxyurea/pharmacology , Minichromosome Maintenance Proteins/genetics , Minichromosome Maintenance Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
11.
Cell Rep ; 5(3): 826-38, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24210823

ABSTRACT

The ubiquitin-modification status of proteins in cells is highly dynamic and maintained by specific ligation machineries (E3 ligases) that tag proteins with ubiquitin or by deubiquitinating enzymes (DUBs) that remove the ubiquitin tag. The development of tools that offset this balance is critical in characterizing signaling pathways that utilize such ubiquitination switches. Herein, we generated a DUB-resistant ubiquitin mutant that is recalcitrant to cleavage by various families of DUBs both in vitro and in mammalian cells. As a proof-of-principle experiment, ectopic expression of the uncleavable ubiquitin stabilized monoubiquitinated PCNA in the absence of DNA damage and also revealed a defect in the clearance of the DNA damage response at unprotected telomeres. Importantly, a proteomic survey using the uncleavable ubiquitin identified ubiquitinated substrates, validating the DUB-resistant ubiquitin expression system as a valuable tool for interrogating cell signaling pathways.


Subject(s)
Ubiquitin/metabolism , Amino Acid Sequence , Cell Line, Tumor , DNA Damage , HEK293 Cells , Humans , Molecular Sequence Data , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitin/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...