Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 176(1-2): 198-212.e15, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30503211

ABSTRACT

Understanding transcription factor navigation through the nucleus remains critical for developing targeted therapeutics. The GLI1 transcription factor must maintain maximal Hedgehog pathway output in basal cell carcinomas (BCCs), and we have previously shown that resistant BCCs increase GLI1 deacetylation through atypical protein kinase Cι/λ (aPKC) and HDAC1. Here we identify a lamina-associated polypeptide 2 (LAP2) isoform-dependent nuclear chaperoning system that regulates GLI1 movement between the nuclear lamina and nucleoplasm to achieve maximal activation. LAP2ß forms a two-site interaction with the GLI1 zinc-finger domain and acetylation site, stabilizing an acetylation-dependent reserve on the inner nuclear membrane (INM). By contrast, the nucleoplasmic LAP2α competes with LAP2ß for GLI1 while scaffolding HDAC1 to deacetylate the secondary binding site. aPKC functions to promote GLI1 association with LAP2α, promoting egress off the INM. GLI1 intranuclear trafficking by LAP2 isoforms represents a powerful signal amplifier in BCCs with implications for zinc finger-based signal transduction and therapeutics.


Subject(s)
DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Zinc Finger Protein GLI1/metabolism , 3T3 Cells , Animals , Carcinoma, Basal Cell/metabolism , Cell Line , Chromatin , DNA-Binding Proteins/physiology , HEK293 Cells , Hedgehog Proteins/metabolism , Hedgehog Proteins/physiology , Histone Deacetylase 1/metabolism , Humans , Membrane Proteins/physiology , Mice , Molecular Chaperones/metabolism , Nuclear Lamina/metabolism , Nuclear Proteins/metabolism , Protein Isoforms/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcription Factors/metabolism , Zinc Finger Protein GLI1/physiology , Zinc Fingers
2.
Nature ; 606(7912): 188-196, 2022 06.
Article in English | MEDLINE | ID: mdl-35585237

ABSTRACT

Proper ectodermal patterning during human development requires previously identified transcription factors such as GATA3 and p63, as well as positional signalling from regional mesoderm1-6. However, the mechanism by which ectoderm and mesoderm factors act to stably pattern gene expression and lineage commitment remains unclear. Here we identify the protein Gibbin, encoded by the Xia-Gibbs AT-hook DNA-binding-motif-containing 1 (AHDC1) disease gene7-9, as a key regulator of early epithelial morphogenesis. We find that enhancer- or promoter-bound Gibbin interacts with dozens of sequence-specific zinc-finger transcription factors and methyl-CpG-binding proteins to regulate the expression of mesoderm genes. The loss of Gibbin causes an increase in DNA methylation at GATA3-dependent mesodermal genes, resulting in a loss of signalling between developing dermal and epidermal cell types. Notably, Gibbin-mutant human embryonic stem-cell-derived skin organoids lack dermal maturation, resulting in p63-expressing basal cells that possess defective keratinocyte stratification. In vivo chimeric CRISPR mouse mutants reveal a spectrum of Gibbin-dependent developmental patterning defects affecting craniofacial structure, abdominal wall closure and epidermal stratification that mirror patient phenotypes. Our results indicate that the patterning phenotypes seen in Xia-Gibbs and related syndromes derive from abnormal mesoderm maturation as a result of gene-specific DNA methylation decisions.


Subject(s)
DNA-Binding Proteins , Epithelium , Gene Expression Regulation, Developmental , Mesoderm , Morphogenesis , Animals , Humans , Mice , Dermis/cytology , Dermis/embryology , Dermis/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Ectoderm/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epidermal Cells/cytology , Epidermal Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/embryology , GATA3 Transcription Factor , Mesoderm/metabolism , Mutation , Organoids , Trans-Activators , Transcription Factors/metabolism
3.
Cell ; 144(3): 319-21, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21295692

ABSTRACT

Directed cell migration polarizes the cytoskeleton, allowing the cell to move toward migratory cues. In this issue, Wu et al. (2011) demonstrate that the glycogen synthase kinase 3ß (GSK3ß) controls microtubule architecture and polarized movement of skin stem cells during wound healing in mammals by regulating the microtubule crosslinking protein ACF7.

4.
PLoS Comput Biol ; 17(9): e1009382, 2021 09.
Article in English | MEDLINE | ID: mdl-34543288

ABSTRACT

The repurposing of biomedical data is inhibited by its fragmented and multi-formatted nature that requires redundant investment of time and resources by data scientists. This is particularly true for Type 1 Diabetes (T1D), one of the most intensely studied common childhood diseases. Intense investigation of the contribution of pancreatic ß-islet and T-lymphocytes in T1D has been made. However, genetic contributions from B-lymphocytes, which are known to play a role in a subset of T1D patients, remain relatively understudied. We have addressed this issue through the creation of Biomedical Data Commons (BMDC), a knowledge graph that integrates data from multiple sources into a single queryable format. This increases the speed of analysis by multiple orders of magnitude. We develop a pipeline using B-lymphocyte multi-dimensional epigenome and connectome data and deploy BMDC to assess genetic variants in the context of Type 1 Diabetes (T1D). Pipeline-identified variants are primarily common, non-coding, poorly conserved, and are of unknown clinical significance. While variants and their chromatin connectivity are cell-type specific, they are associated with well-studied disease genes in T-lymphocytes. Candidates include established variants in the HLA-DQB1 and HLA-DRB1 and IL2RA loci that have previously been demonstrated to protect against T1D in humans and mice providing validation for this method. Others are included in the well-established T1D GRS2 genetic risk scoring method. More intriguingly, other prioritized variants are completely novel and form the basis for future mechanistic and clinical validation studies The BMDC community-based platform can be expanded and repurposed to increase the accessibility, reproducibility, and productivity of biomedical information for diverse applications including the prioritization of cell type-specific disease alleles from complex phenotypes.


Subject(s)
B-Lymphocytes/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Animals , Child , Computational Biology , Databases, Genetic/statistics & numerical data , Gene Regulatory Networks , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study/statistics & numerical data , HLA-DQ beta-Chains/genetics , HLA-DRB1 Chains/genetics , Humans , Ikaros Transcription Factor/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Mice , Polymorphism, Single Nucleotide , RNA, Untranslated/genetics
5.
Cell ; 146(2): 334-334.e2, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21784251
6.
Proc Natl Acad Sci U S A ; 115(52): E12407-E12416, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30530649

ABSTRACT

The genetically heterogeneous spinocerebellar ataxias (SCAs) are caused by Purkinje neuron dysfunction and degeneration, but their underlying pathological mechanisms remain elusive. The Src family of nonreceptor tyrosine kinases (SFK) are essential for nervous system homeostasis and are increasingly implicated in degenerative disease. Here we reveal that the SFK suppressor Missing-in-metastasis (MTSS1) is an ataxia locus that links multiple SCAs. MTSS1 loss results in increased SFK activity, reduced Purkinje neuron arborization, and low basal firing rates, followed by cell death. Surprisingly, mouse models for SCA1, SCA2, and SCA5 show elevated SFK activity, with SCA1 and SCA2 displaying dramatically reduced MTSS1 protein levels through reduced gene expression and protein translation, respectively. Treatment of each SCA model with a clinically approved Src inhibitor corrects Purkinje neuron basal firing and delays ataxia progression in MTSS1 mutants. Our results identify a common SCA therapeutic target and demonstrate a key role for MTSS1/SFK in Purkinje neuron survival and ataxia progression.


Subject(s)
Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/physiopathology , Animals , Ataxia/pathology , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , Proteins/metabolism , Purkinje Cells/physiology , Spinocerebellar Ataxias/metabolism , Spinocerebellar Degenerations/metabolism , Spinocerebellar Degenerations/physiopathology , src-Family Kinases/metabolism
7.
Genes Dev ; 27(11): 1217-22, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23752588

ABSTRACT

The signals regulating stem cell activation during tissue regeneration remain poorly understood. We investigated the baldness associated with mutations in the voltage-gated calcium channel (VGCC) Cav1.2 underlying Timothy syndrome (TS). While hair follicle stem cells express Cav1.2, they lack detectable voltage-dependent calcium currents. Cav1.2(TS) acts in a dominant-negative manner to markedly delay anagen, while L-type channel blockers act through Cav1.2 to induce anagen and overcome the TS phenotype. Cav1.2 regulates production of the bulge-derived BMP inhibitor follistatin-like1 (Fstl1), derepressing stem cell quiescence. Our findings show how channels act in nonexcitable tissues to regulate stem cells and may lead to novel therapeutics for tissue regeneration.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling , Calcium/metabolism , Hair Follicle/cytology , Stem Cells/cytology , Stem Cells/metabolism , Animals , Autistic Disorder , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Calcium Signaling/drug effects , Follistatin-Related Proteins/biosynthesis , Follistatin-Related Proteins/metabolism , Long QT Syndrome/metabolism , Mice , Stem Cells/drug effects , Syndactyly/metabolism
8.
Genes Dev ; 26(11): 1235-46, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22661232

ABSTRACT

During hair follicle morphogenesis, dermal papillae (DPs) function as mesenchymal signaling centers that cross-talk with overlying epithelium to regulate morphogenesis. While the DP regulates hair follicle formation, relatively little is known about the molecular basis of DP formation. The morphogen Sonic hedgehog (Shh) is known for regulating hair follicle epithelial growth, with excessive signaling resulting in basal cell carcinomas. Here, we investigate how dermal-specific Shh signaling contributes to DP formation and hair growth. Using a Cre-lox genetic model and RNAi in hair follicle reconstitution assays, we demonstrate that dermal Smoothened (Smo) loss of function results in the loss of the DP precursor, the dermal condensate, and a stage 2 hair follicle arrest phenotype reminiscent of Shh(-/-) skin. Surprisingly, dermal Smo does not regulate cell survival or epithelial proliferation. Rather, molecular screening and immunostaining studies reveal that dermal Shh signaling controls the expression of a subset of DP-specific signature genes. Using a hairpin/cDNA lentiviral system, we show that overexpression of the Shh-dependent gene Noggin, but not Sox2 or Sox18, can partially rescue the dermal Smo knockdown hair follicle phenotype by increasing the expression of epithelial Shh. Our findings suggest that dermal Shh signaling regulates specific DP signatures to maintain DP maturation while maintaining a reciprocal Shh-Noggin signaling loop to drive hair follicle morphogenesis.


Subject(s)
Hair Follicle/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Skin/cytology , Skin/metabolism , Animals , Female , Gene Knockdown Techniques , Hair/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Receptors, G-Protein-Coupled/genetics , Smoothened Receptor
9.
Nature ; 494(7438): 484-8, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23446420

ABSTRACT

Growth of basal cell carcinomas (BCCs) requires high levels of hedgehog (HH) signalling through the transcription factor GLI. Although inhibitors of membrane protein smoothened (SMO) effectively suppress HH signalling, early tumour resistance illustrates the need for additional downstream targets for therapy. Here we identify atypical protein kinase C ι/λ (aPKC-ι/λ) as a novel GLI regulator in mammals. aPKC-ι/λ and its polarity signalling partners co-localize at the centrosome and form a complex with missing-in-metastasis (MIM), a scaffolding protein that potentiates HH signalling. Genetic or pharmacological loss of aPKC-ι/λ function blocks HH signalling and proliferation of BCC cells. Prkci is a HH target gene that forms a positive feedback loop with GLI and exists at increased levels in BCCs. Genome-wide transcriptional profiling shows that aPKC-ι/λ and SMO control the expression of similar genes in tumour cells. aPKC-ι/λ functions downstream of SMO to phosphorylate and activate GLI1, resulting in maximal DNA binding and transcriptional activation. Activated aPKC-ι/λ is upregulated in SMO-inhibitor-resistant tumours and targeting aPKC-ι/λ suppresses signalling and growth of resistant BCC cell lines. These results demonstrate that aPKC-ι/λ is critical for HH-dependent processes and implicates aPKC-ι/λ as a new, tumour-selective therapeutic target for the treatment of SMO-inhibitor-resistant cancers.


Subject(s)
Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/pathology , Isoenzymes/metabolism , Kruppel-Like Transcription Factors/metabolism , Protein Kinase C/metabolism , Transcription Factors/metabolism , Animals , Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Centrosome/metabolism , Drug Resistance, Neoplasm , Feedback, Physiological , Hedgehog Proteins/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Keratinocytes/metabolism , Kruppel-Like Transcription Factors/genetics , Mice , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Smoothened Receptor , Zinc Finger Protein GLI1
10.
Genes Dev ; 25(22): 2333-46, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22051878

ABSTRACT

The Hedgehog (Hh) pathway is essential for vertebrate embryogenesis, and excessive Hh target gene activation can cause cancer in humans. Here we show that Neuropilin 1 (Nrp1) and Nrp2, transmembrane proteins with roles in axon guidance and vascular endothelial growth factor (VEGF) signaling, are important positive regulators of Hh signal transduction. Nrps are expressed at times and locations of active Hh signal transduction during mouse development. Using cell lines lacking key Hh pathway components, we show that Nrps mediate Hh transduction between activated Smoothened (Smo) protein and the negative regulator Suppressor of Fused (SuFu). Nrp1 transcription is induced by Hh signaling, and Nrp1 overexpression increases maximal Hh target gene activation, indicating the existence of a positive feedback circuit. The regulation of Hh signal transduction by Nrps is conserved between mammals and bony fish, as we show that morpholinos targeting the Nrp zebrafish ortholog nrp1a produce a specific and highly penetrant Hh pathway loss-of-function phenotype. These findings enhance our knowledge of Hh pathway regulation and provide evidence for a conserved nexus between Nrps and this important developmental signaling system.


Subject(s)
Hedgehog Proteins/metabolism , Neuropilins/metabolism , Signal Transduction , Animals , Feedback, Physiological , Gene Expression Regulation, Developmental , Mice , Neuropilin-1/genetics , Neuropilin-1/metabolism , Neuropilin-2/genetics , Neuropilin-2/metabolism , RNA Interference , Receptors, G-Protein-Coupled/metabolism , Repressor Proteins/metabolism , Smoothened Receptor
11.
N Engl J Med ; 366(23): 2171-9, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-22670903

ABSTRACT

BACKGROUND: Alterations in hedgehog signaling are implicated in the pathogenesis of basal-cell carcinoma. Although most basal-cell carcinomas are treated surgically, no effective therapy exists for locally advanced or metastatic basal-cell carcinoma. A phase 1 study of vismodegib (GDC-0449), a first-in-class, small-molecule inhibitor of the hedgehog pathway, showed a 58% response rate among patients with advanced basal-cell carcinoma. METHODS: In this multicenter, international, two-cohort, nonrandomized study, we enrolled patients with metastatic basal-cell carcinoma and those with locally advanced basal-cell carcinoma who had inoperable disease or for whom surgery was inappropriate (because of multiple recurrences and a low likelihood of surgical cure, or substantial anticipated disfigurement). All patients received 150 mg of oral vismodegib daily. The primary end point was the independently assessed objective response rate; the primary hypotheses were that the response rate would be greater than 20% for patients with locally advanced basal-cell carcinoma and greater than 10% for those with metastatic basal-cell carcinoma. RESULTS: In 33 patients with metastatic basal-cell carcinoma, the independently assessed response rate was 30% (95% confidence interval [CI], 16 to 48; P=0.001). In 63 patients with locally advanced basal-cell carcinoma, the independently assessed response rate was 43% (95% CI, 31 to 56; P<0.001), with complete responses in 13 patients (21%). The median duration of response was 7.6 months in both cohorts. Adverse events occurring in more than 30% of patients were muscle spasms, alopecia, dysgeusia (taste disturbance), weight loss, and fatigue. Serious adverse events were reported in 25% of patients; seven deaths due to adverse events were noted. CONCLUSIONS: Vismodegib is associated with tumor responses in patients with locally advanced or metastatic basal-cell carcinoma. (Funded by Genentech; Erivance BCC ClinicalTrials.gov number, NCT00833417.).


Subject(s)
Anilides/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Basal Cell/drug therapy , Hedgehog Proteins/antagonists & inhibitors , Pyridines/therapeutic use , Skin Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Aged, 80 and over , Anilides/adverse effects , Antineoplastic Agents/adverse effects , Carcinoma, Basal Cell/pathology , Carcinoma, Basal Cell/secondary , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Pyridines/adverse effects , Signal Transduction/drug effects , Skin Neoplasms/pathology , Treatment Outcome
12.
Stem Cells ; 32(1): 85-92, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23963711

ABSTRACT

The activation of tissue stem cells from their quiescent state represents the initial step in the complex process of organ regeneration and tissue repair. While the identity and location of tissue stem cells are becoming known, how key regulators control the balance of activation and quiescence remains mysterious. The vertebrate hair is an ideal model system where hair cycling between growth and resting phases is precisely regulated by morphogen signaling pathways, but how these events are coordinated to promote orderly signaling in a spatial and temporal manner remains unclear. Here, we show that hair cycle timing depends on regulated stability of signaling substrates by the ubiquitin-proteasome system. Topical application of partial proteasomal inhibitors (PaPIs) inhibits epidermal and dermal proteasome activity throughout the hair cycle. PaPIs prevent the destruction of the key anagen signal ß-catenin, resulting in more rapid hair growth and dramatically shortened telogen. We show that PaPIs induce excess ß-catenin, act similarly to the GSK3ß antagonist LiCl, and antagonize Dickopf-related protein-mediated inhibition of anagen. PaPIs thus represent a novel class of hair growth agents that act through transiently modifying the balance of stem cell activation and quiescence pathways.


Subject(s)
Hair Follicle/drug effects , Hair Follicle/growth & development , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , beta Catenin/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Female , Hair Follicle/cytology , Hair Follicle/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism
13.
Mol Ther ; 22(4): 725-33, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24390279

ABSTRACT

Definitive correction of disease causing mutations in somatic cells by homologous recombination (HR) is an attractive therapeutic approach for the treatment of genetic diseases. However, HR-based somatic gene therapy is limited by the low efficiency of gene targeting in mammalian cells and replicative senescence of primary cells ex vivo, forcing investigators to explore alternative strategies such as retro- and lentiviral gene transfer, or genome editing in induced pluripotent stem cells. Here, we report correction of mutations at the LAMA3 locus in primary keratinocytes derived from a patient affected by recessive inherited Herlitz junctional epidermolysis bullosa (H-JEB) disorder using recombinant adenoassociated virus (rAAV)-mediated HR. We identified a highly recombinogenic AAV serotype, AAV-DJ, that mediates efficient gene targeting in keratinocytes at clinically relevant frequencies with a low rate of random integration. Targeted H-JEB patient cells were selected based on restoration of adhesion phenotype, which eliminated the need for foreign sequences in repaired cells, enhancing the clinical use and safety profile of our approach. Corrected pools of primary cells assembled functional laminin-332 heterotrimer and fully reversed the blistering phenotype both in vitro and in skin grafts. The efficient targeting of the LAMA3 locus by AAV-DJ using phenotypic selection, together with the observed low frequency of off-target events, makes AAV-DJ based somatic cell targeting a promising strategy for ex vivo therapy for this severe and often lethal epithelial disorder.


Subject(s)
Epidermolysis Bullosa, Junctional/genetics , Genetic Therapy/methods , Homologous Recombination/genetics , Laminin/genetics , Animals , Collagen Type VII/genetics , Dependovirus/genetics , Epidermolysis Bullosa, Junctional/pathology , Epidermolysis Bullosa, Junctional/therapy , Heterografts , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mutation
14.
Curr Opin Cell Biol ; 19(6): 691-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18061425

ABSTRACT

Intense focus has been centered around how the primary cilia transduces the hedgehog (Hh) signal from smoothened (Smo) to the Gli transcription factors. New data indicate that ligand and signaling lipids help regulate small GTPase-dependent accumulation and activity of signaling components.


Subject(s)
Cilia/physiology , Hedgehog Proteins/metabolism , Monomeric GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Transcription Factors/metabolism , Humans , Smoothened Receptor , Zinc Finger Protein GLI1
15.
J Invest Dermatol ; 144(6): 1368-1377.e6, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38157930

ABSTRACT

Although basal cell carcinomas arise from ectopic Hedgehog pathway activation and can be treated with pathway inhibitors, sporadic basal cell carcinomas display high resistance rates, whereas tumors arising in patients with Gorlin syndrome with germline Patched (PTCH1) alterations are uniformly suppressed by inhibitor therapy. In rare cases, patients with Gorlin syndrome on long-term inhibitor therapy will develop individual resistant tumor clones that rapidly progress, but the basis of this resistance remains unstudied. In this study, we report a case of an SMO inhibitor-resistant tumor arising in a patient with Gorlin syndrome on suppressive SMO inhibitor for nearly a decade. Using a combination of multiomics and spatial transcriptomics, we define the tumor populations at the cellular and tissue level to conclude that Gorlin tumors can develop resistance to SMO inhibitors through the previously described basal to squamous cell carcinoma transition. Intriguingly, through spatial whole-exome genomic analysis, we nominate PCYT2, ETNK1, and the phosphatidylethanolamine biosynthetic pathway as genetic suppressors of basal to squamous cell carcinoma transition resistance. These observations provide a general framework for studying tumor evolution and provide important clinical insight into mechanisms of resistance to SMO inhibitors for not only Gorlin syndrome but also sporadic basal cell carcinomas.


Subject(s)
Basal Cell Nevus Syndrome , Carcinoma, Basal Cell , Carcinoma, Squamous Cell , Drug Resistance, Neoplasm , Skin Neoplasms , Smoothened Receptor , Humans , Basal Cell Nevus Syndrome/genetics , Basal Cell Nevus Syndrome/pathology , Basal Cell Nevus Syndrome/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Smoothened Receptor/genetics , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/pathology , Male , Anilides/therapeutic use , Female , Signal Transduction/drug effects , Pyridines/therapeutic use
16.
Nat Commun ; 15(1): 5834, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992003

ABSTRACT

We present Dystrophic Epidermolysis Bullosa Cell Therapy (DEBCT), a scalable platform producing autologous organotypic iPS cell-derived induced skin composite (iSC) grafts for definitive treatment. Clinical-grade manufacturing integrates CRISPR-mediated genetic correction with reprogramming into one step, accelerating derivation of COL7A1-edited iPS cells from patients. Differentiation into epidermal, dermal and melanocyte progenitors is followed by CD49f-enrichment, minimizing maturation heterogeneity. Mouse xenografting of iSCs from four patients with different mutations demonstrates disease modifying activity at 1 month. Next-generation sequencing, biodistribution and tumorigenicity assays establish a favorable safety profile at 1-9 months. Single cell transcriptomics reveals that iSCs are composed of the major skin cell lineages and include prominent holoclone stem cell-like signatures of keratinocytes, and the recently described Gibbin-dependent signature of fibroblasts. The latter correlates with enhanced graftability of iSCs. In conclusion, DEBCT overcomes manufacturing and safety roadblocks and establishes a reproducible, safe, and cGMP-compatible therapeutic approach to heal lesions of DEB patients.


Subject(s)
Cell- and Tissue-Based Therapy , Collagen Type VII , Epidermolysis Bullosa Dystrophica , Induced Pluripotent Stem Cells , Humans , Epidermolysis Bullosa Dystrophica/therapy , Epidermolysis Bullosa Dystrophica/genetics , Animals , Induced Pluripotent Stem Cells/transplantation , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Mice , Collagen Type VII/genetics , Collagen Type VII/metabolism , Cell- and Tissue-Based Therapy/methods , Fibroblasts/metabolism , Cell Differentiation , Keratinocytes/metabolism , Keratinocytes/transplantation , Skin/metabolism , Transplantation, Autologous , Male , Mutation , Female , Skin Transplantation/methods , Gene Editing/methods , CRISPR-Cas Systems
17.
bioRxiv ; 2023 Jul 27.
Article in English | MEDLINE | ID: mdl-37546976

ABSTRACT

While basal cell carcinomas (BCCs) arise from ectopic hedgehog pathway activation and can be treated with pathway inhibitors, sporadic BCCs display high resistance rates while tumors arising in Gorlin syndrome patients with germline Patched ( PTCH1 ) mutations are uniformly suppressed by inhibitor therapy. In rare cases, Gorlin syndrome patients on long-term inhibitor therapy will develop individual resistant tumor clones that rapidly progress, but the basis of this resistance remains unstudied. Here we report a case of an SMO i -resistant tumor arising in a Gorlin patient on suppressive SMO i for nearly a decade. Using a combination of multi-omics and spatial transcriptomics, we define the tumor populations at the cellular and tissue level to conclude that Gorlin tumors can develop resistance to SMO i through the previously described basal to squamous cell carcinoma transition (BST). Intriguingly, through spatial whole exome genomic analysis, we nominate PCYT2, ETNK1, and the phosphatidylethanolamine biosynthetic pathway as novel genetic suppressors of BST resistance. These observations provide a general framework for studying tumor evolution and provide important clinical insight into mechanisms of resistance to SMO i for not only Gorlin syndrome but sporadic BCCs as well.

18.
Nat Genet ; 55(8): 1288-1300, 2023 08.
Article in English | MEDLINE | ID: mdl-37500727

ABSTRACT

Genome-wide association studies have identified many loci associated with hair and skin disease, but identification of causal variants requires deciphering of gene-regulatory networks in relevant cell types. We generated matched single-cell chromatin profiles and transcriptomes from scalp tissue from healthy controls and patients with alopecia areata, identifying diverse cell types of the hair follicle niche. By interrogating these datasets at multiple levels of cellular resolution, we infer 50-100% more enhancer-gene links than previous approaches and show that aggregate enhancer accessibility for highly regulated genes predicts expression. We use these gene-regulatory maps to prioritize cell types, genes and causal variants implicated in the pathobiology of androgenetic alopecia (AGA), eczema and other complex traits. AGA genome-wide association studies signals are enriched in dermal papilla regulatory regions, supporting the role of these cells as drivers of AGA pathogenesis. Finally, we train machine learning models to nominate single-nucleotide polymorphisms that affect gene expression through disruption of transcription factor binding, predicting candidate functional single-nucleotide polymorphism for AGA and eczema.


Subject(s)
Alopecia Areata , Eczema , Humans , Scalp/metabolism , Chromatin/genetics , Chromatin/metabolism , Genome-Wide Association Study , Transcriptome/genetics , Alopecia Areata/metabolism , Hair Follicle/metabolism , Eczema/genetics , Eczema/metabolism
19.
iScience ; 26(3): 106125, 2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36843855

ABSTRACT

Ectodermal dysplasias including skin abnormalities and cleft lip/palate result from improper surface ectoderm (SE) patterning. However, the connection between SE gene regulatory networks and disease remains poorly understood. Here, we dissect human SE differentiation with multiomics and establish GRHL2 as a key mediator of early SE commitment, which acts by skewing cell fate away from the neural lineage. GRHL2 and master SE regulator AP2a balance early cell fate output, with GRHL2 facilitating AP2a binding to SE loci. In turn, AP2a restricts GRHL2 DNA binding away from de novo chromatin contacts. Integration of these regulatory sites with ectodermal dysplasia-associated genomic variants annotated within the Biomedical Data Commons identifies 55 loci previously implicated in craniofacial disorders. These include ABCA4/ARHGAP29 and NOG regulatory regions where disease-linked variants directly affect GRHL2/AP2a binding and gene transcription. These studies elucidate the logic underlying SE commitment and deepen our understanding of human oligogenic disease pathogenesis.

20.
Nat Commun ; 14(1): 2685, 2023 05 10.
Article in English | MEDLINE | ID: mdl-37164949

ABSTRACT

Cancer immunotherapies have revolutionized treatment but have shown limited success as single-agent therapies highlighting the need to understand the origin, assembly, and dynamics of heterogeneous tumor immune niches. Here, we use single-cell and imaging-based spatial analysis to elucidate three microenvironmental neighborhoods surrounding the heterogeneous basal cell carcinoma tumor epithelia. Within the highly proliferative neighborhood, we find that TREM2+ skin cancer-associated macrophages (SCAMs) support the proliferation of a distinct tumor epithelial population through an immunosuppression-independent manner via oncostatin-M/JAK-STAT3 signaling. SCAMs represent a unique tumor-specific TREM2+ population defined by VCAM1 surface expression that is not found in normal homeostatic skin or during wound healing. Furthermore, SCAMs actively proliferate and self-propagate through multiple serial tumor passages, indicating long-term potential. The tumor rapidly drives SCAM differentiation, with intratumoral injections sufficient to instruct naive bone marrow-derived monocytes to polarize within days. This work provides mechanistic insights into direct tumor-immune niche dynamics independent of immunosuppression, providing the basis for potential combination tumor therapies.


Subject(s)
Carcinoma, Basal Cell , Skin Neoplasms , Humans , Macrophages/metabolism , Monocytes , Carcinogenesis/metabolism , Carcinoma, Basal Cell/metabolism , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL