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

Country/Region as subject
Publication year range
1.
EMBO Rep ; 22(3): e52152, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33554445

ABSTRACT

"Flash forward genetics" refers to a genetic approach based on the functional interaction of a given factor with unknown partner(s) converging on shared targets across evolutionary boundaries. A study by Li et al (2021), published in this issue of EMBO Reports, illustrates the innovative potential of the approach. The authors applied it to identify interacting factors for FOXN1, a mammalian transcription factor with a highly specialized function in hair follicle morphogenesis and thymus. The authors express FOXN1 in the Drosophila eye to perform an unbiased genetic screen in a totally heterologous system. In a remarkable tour de force, the authors identify and characterize a factor so far known for its ubiquitous function in transcription elongation, AFF4. Li et al show that AFF4 plays also a specific role in hair follicle and thymus development in the mouse overlapping with that of FOXN1.


Subject(s)
Forkhead Transcription Factors , Hair Follicle , Animals , Biological Evolution , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Hair Follicle/metabolism , Mice , Mice, Nude
2.
Nucleic Acids Res ; 49(7): 3748-3763, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33764436

ABSTRACT

Epigenetic regulation of cell and tissue function requires the coordinated action of transcription factors. However, their combinatorial activities during regeneration remain largely unexplored. Here, we discover an unexpected interaction between the cytoprotective transcription factor NRF2 and p63- a key player in epithelial morphogenesis. Chromatin immunoprecipitation combined with sequencing and reporter assays identifies enhancers and promoters that are simultaneously activated by NRF2 and p63 in human keratinocytes. Modeling of p63 and NRF2 binding to nucleosomal DNA suggests their chromatin-assisted interaction. Pharmacological and genetic activation of NRF2 increases NRF2-p63 binding to enhancers and promotes keratinocyte proliferation, which involves the common NRF2-p63 target cyclin-dependent kinase 12. These results unravel a collaborative function of NRF2 and p63 in the control of epidermal renewal and suggest their combined activation as a strategy to promote repair of human skin and other stratified epithelia.


Subject(s)
Keratinocytes , NF-E2-Related Factor 2/physiology , Skin , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinases/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Skin/cytology , Skin/metabolism
3.
Proc Natl Acad Sci U S A ; 115(5): E906-E915, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29339502

ABSTRACT

The p63 gene encodes a master regulator of epidermal commitment, development, and differentiation. Heterozygous mutations in the C-terminal domain of the p63 gene can cause ankyloblepharon-ectodermal defects-cleft lip/palate (AEC) syndrome, a life-threatening disorder characterized by skin fragility and severe, long-lasting skin erosions. Despite deep knowledge of p63 functions, little is known about mechanisms underlying disease pathology and possible treatments. Here, we show that multiple AEC-associated p63 mutations, but not those causative of other diseases, lead to thermodynamic protein destabilization, misfolding, and aggregation, similar to the known p53 gain-of-function mutants found in cancer. AEC mutant proteins exhibit impaired DNA binding and transcriptional activity, leading to dominant negative effects due to coaggregation with wild-type p63 and p73. Importantly, p63 aggregation occurs also in a conditional knock-in mouse model for the disorder, in which the misfolded p63 mutant protein leads to severe epidermal defects. Variants of p63 that abolish aggregation of the mutant proteins are able to rescue p63's transcriptional function in reporter assays as well as in a human fibroblast-to-keratinocyte conversion assay. Our studies reveal that AEC syndrome is a protein aggregation disorder and opens avenues for therapeutic intervention.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Eye Abnormalities/genetics , Phosphoproteins/genetics , Skin/pathology , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Animals , Ectoderm/metabolism , Frameshift Mutation , HEK293 Cells , Heterozygote , Humans , Mice , Mutation , Mutation, Missense , Protein Binding , Protein Denaturation , Transcription, Genetic
4.
PLoS Genet ; 13(6): e1006828, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28604778

ABSTRACT

Cleft palate is a common congenital disorder that affects up to 1 in 2500 live births and results in considerable morbidity to affected individuals and their families. The aetiology of cleft palate is complex with both genetic and environmental factors implicated. Mutations in the transcription factor p63 are one of the major individual causes of cleft palate; however, the gene regulatory networks in which p63 functions remain only partially characterized. Our findings demonstrate that p63 functions as an essential regulatory molecule in the spatio-temporal control of palatal epithelial cell fate to ensure appropriate fusion of the palatal shelves. Initially, p63 induces periderm formation and controls its subsequent maintenance to prevent premature adhesion between adhesion-competent, intra-oral epithelia. Subsequently, TGFß3-induced down-regulation of p63 in the medial edge epithelia of the palatal shelves is a pre-requisite for palatal fusion by facilitating periderm migration from, and reducing the proliferative potential of, the midline epithelial seam thereby preventing cleft palate.


Subject(s)
Cleft Palate/genetics , Gene Regulatory Networks/genetics , Phosphoproteins/genetics , Trans-Activators/genetics , Transforming Growth Factor beta3/genetics , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Cleft Palate/physiopathology , Disease Models, Animal , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Mutation , Phosphoproteins/biosynthesis , Signal Transduction/genetics , Trans-Activators/biosynthesis
5.
Int J Mol Sci ; 21(2)2020 Jan 17.
Article in English | MEDLINE | ID: mdl-31963474

ABSTRACT

Early events of basal cell carcinoma (BCC) tumorigenesis are triggered by inappropriate activation of SHH signaling, via the loss of Patched1 (Ptch1) or by activating mutations of Smoothened (Smo). TBX1 is a key regulator of pharyngeal development, mainly through expression in multipotent progenitor cells of the cardiopharyngeal lineage. This transcription factor is connected to several major signaling systems, such as FGF, WNT, and SHH, and it has been linked to cell proliferation and to the regulation of cell shape and cell dynamics. Here, we show that TBX1 was expressed in all of the 51 BCC samples that we have tested, while in healthy human skin it was only expressed in the hair follicle. Signal intensity and distribution was heterogeneous among tumor samples. Experiments performed on a cellular model of mouse BCC showed that Tbx1 is downstream to GLI2, a factor in the SHH signaling, and that, in turn, it regulates the expression of Dvl2, which encodes an adaptor protein that is necessary for the transduction of WNT signaling. Consistently, Tbx1 depletion in the cellular model significantly reduced cell migration. These results suggest that TBX1 is part of a core transcription network that promotes BCC tumorigenesis.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Basal Cell/pathology , Dishevelled Proteins/metabolism , Nuclear Proteins/metabolism , Skin Neoplasms/pathology , T-Box Domain Proteins/metabolism , Zinc Finger Protein Gli2/metabolism , Adult , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/genetics , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/metabolism , Case-Control Studies , Cell Proliferation , Dishevelled Proteins/genetics , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , Nuclear Proteins/genetics , Prognosis , Retrospective Studies , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , T-Box Domain Proteins/genetics , Tumor Cells, Cultured , Zinc Finger Protein Gli2/genetics
6.
J Investig Dermatol Symp Proc ; 19(2): S98-S100, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30471766

ABSTRACT

The p53 family member p63 is a master regulator of gene expression in stratified epithelia, such as the epidermis. One of the main functions of p63 is to sustain mechanical resistance, positively regulating several epidermal genes involved in cell-matrix adhesion and cell-cell adhesion (Ferone et al., 2015).

7.
Nucleic Acids Res ; 43(2): 862-74, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25567987

ABSTRACT

p63 is a crucial regulator of epidermal development, but its transcriptional control has remained elusive. Here, we report the identification of a long-range enhancer (p63LRE) that is composed of two evolutionary conserved modules (C38 and C40), acting in concert to control tissue- and layer-specific expression of the p63 gene. Both modules are in an open and active chromatin state in human and mouse keratinocytes and in embryonic epidermis, and are strongly bound by p63. p63LRE activity is dependent on p63 expression in embryonic skin, and also in the commitment of human induced pluripotent stem cells toward an epithelial cell fate. A search for other transcription factors involved in p63LRE regulation revealed that the CAAT enhancer binding proteins Cebpa and Cebpb and the POU domain-containing protein Pou3f1 repress p63 expression during keratinocyte differentiation by binding the p63LRE enhancer. Collectively, our data indicate that p63LRE is composed of additive and partly redundant enhancer modules that act to direct robust p63 expression selectively in the basal layer of the epidermis.


Subject(s)
Enhancer Elements, Genetic , Epidermis/embryology , Epidermis/metabolism , Gene Expression Regulation, Developmental , Keratinocytes/metabolism , Phosphoproteins/genetics , Trans-Activators/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Humans , Keratinocytes/cytology , Mice, Inbred C57BL , Morphogenesis/genetics , Transcription Factors/metabolism , Transcriptional Activation
8.
Hum Mol Genet ; 22(3): 531-43, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23108156

ABSTRACT

Ankyloblepharon, ectodermal defects, cleft lip/palate (AEC) syndrome is a rare autosomal dominant disorder caused by mutations in the p63 gene, essential for embryonic development of stratified epithelia. The most severe cutaneous manifestation of this disorder is the long-lasting skin fragility associated with severe skin erosions after birth. Using a knock-in mouse model for AEC syndrome, we found that skin fragility was associated with microscopic blistering between the basal and suprabasal compartments of the epidermis and reduced desmosomal contacts. Expression of desmosomal cadherins and desmoplakin was strongly reduced in AEC mutant keratinocytes and in newborn epidermis. A similar impairment in desmosome gene expression was observed in human keratinocytes isolated from AEC patients, in p63-depleted keratinocytes and in p63 null embryonic skin, indicating that p63 mutations causative of AEC syndrome have a dominant-negative effect on the wild-type p63 protein. Among the desmosomal components, desmocollin 3, desmoplakin and desmoglein 1 were the most significantly reduced by mutant p63 both at the RNA and protein levels. Chromatin immunoprecipitation experiments and transactivation assays revealed that p63 controls these genes at the transcriptional level. Consistent with reduced desmosome function, AEC mutant and p63-deficient keratinocytes had an impaired ability to withstand mechanical stress, which was alleviated by epidermal growth factor receptor inhibitors known to stabilize desmosomes. Our study reveals that p63 is a crucial regulator of a subset of desmosomal genes and that this function is impaired in AEC syndrome. Reduced mechanical strength resulting from p63 mutations can be alleviated pharmacologically by increasing desmosome adhesion with possible therapeutic implications.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Desmosomes/metabolism , Ectodermal Dysplasia/genetics , Eye Abnormalities/genetics , Membrane Proteins/metabolism , Animals , Cell Adhesion , Cells, Cultured , Chromatin Immunoprecipitation , Cleft Lip/pathology , Cleft Palate/pathology , Cloning, Molecular , Desmosomes/genetics , Ectodermal Dysplasia/pathology , Epidermis/metabolism , Epidermis/physiopathology , Epithelium/metabolism , Epithelium/physiopathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Eye Abnormalities/pathology , Eyelids/abnormalities , Eyelids/pathology , Female , Gene Expression Regulation , Humans , Keratinocytes/metabolism , Luciferases/analysis , Luciferases/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Real-Time Polymerase Chain Reaction , Skin/metabolism , Skin/physiopathology
9.
Am J Hum Genet ; 90(1): 69-75, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22197488

ABSTRACT

Pterygium syndromes are complex congenital disorders that encompass several distinct clinical conditions characterized by multiple skin webs affecting the flexural surfaces often accompanied by craniofacial anomalies. In severe forms, such as in the autosomal-recessive Bartsocas-Papas syndrome, early lethality is common, complicating the identification of causative mutations. Using exome sequencing in a consanguineous family, we identified the homozygous mutation c.1127C>A in exon 7 of RIPK4 that resulted in the introduction of the nonsense mutation p.Ser376X into the encoded ankyrin repeat-containing kinase, a protein that is essential for keratinocyte differentiation. Subsequently, we identified a second mutation in exon 2 of RIPK4 (c.242T>A) that resulted in the missense variant p.Ile81Asn in the kinase domain of the protein. We have further demonstrated that RIPK4 is a direct transcriptional target of the protein p63, a master regulator of stratified epithelial development, which acts as a nodal point in the cascade of molecular events that prevent pterygium syndromes.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Exome , Protein Serine-Threonine Kinases/genetics , Pterygium/congenital , Amino Acid Sequence , Animals , Base Sequence , Child , Cleft Lip/diagnosis , Cleft Palate/diagnosis , Consanguinity , Craniofacial Abnormalities/genetics , Exons , Genes, Recessive , Genetic Loci , Humans , Keratinocytes/metabolism , Male , Mice , Molecular Sequence Data , Mutation , Phosphoproteins/metabolism , Pterygium/diagnosis , Pterygium/genetics , Severity of Illness Index , Skin Abnormalities , Trans-Activators/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
10.
Cell Tissue Res ; 360(3): 513-28, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25645146

ABSTRACT

As the outermost tissue of the body, the epidermis is the first physical barrier for any pressure, stress or trauma. Several specialized cell-matrix and cell-cell adhesion structures, together with an intracellular network of dedicated intermediate filaments, are required to confer critical resilience to mechanical stress. The transcription factor p63 is a master regulator of gene expression in the epidermis and in other stratified epithelia. It has been extensively demonstrated that p63 positively controls a large number of tissue-specific genes, including those encoding a large fraction of tissue-restricted cell adhesion molecules. Consistent with p63 functions in cell adhesion and in epidermal differentiation, heterozygous mutations clustered mainly in the p63 C-terminus are causative of AEC syndrome, an autosomal dominant disorder characterized by cleft palate, ankyloblepharon and ectodermal dysplasia associated with severe skin erosions, bleeding and infections. The molecular basis of skin erosions in AEC patients is not fully understood, although defects in desmosomes and in other cell junctions are likely to be involved. Here, we provide an extensive review of the different epidermal cell junctions that cooperate to withstand mechanical stress and on the mechanisms by which p63 regulates gene expression of their components in healthy skin and in AEC syndrome. Collectively, advancement in understanding the molecular mechanisms by which epidermal cell junctions precisely exert their functions and how p63 orchestrates their coordinated expression, will ultimately lead to insight into developing future strategies for the treatment of AEC syndrome and more in generally for diseases that share an overlapping phenotype.


Subject(s)
Disease , Epidermis/metabolism , Health , Intercellular Junctions/metabolism , Transcription Factors/metabolism , Animals , Cell-Matrix Junctions/metabolism , Humans , Transcription Factors/chemistry , Transcription Factors/genetics
11.
Exp Dermatol ; 24(2): 114-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25387952

ABSTRACT

Nectins are immunoglobulin-like cell adhesion molecules mainly localized in adherens junctions. The transcription factor p63 is a master regulator of gene expression in stratified epithelia and controls several molecular processes. As mutations in the Pvrl1 and Pvrl4 genes encoding for nectins cause genetic disorders with phenotypes similar to p63-related syndromes, we investigated whether these proteins might be under p63 transcriptional control. Here, we show that in p63-null skin, Pvrl1 gene expression is strongly reduced, whereas Pvrl4 expression is unaffected. In human and mouse primary keratinocytes p63 depletion leads to a specific downregulation of the Pvrl1 gene. Consistent with a direct regulation, chromatin immunoprecipitation experiments (ChIP) indicate that p63 binds to two conserved intronic Pvrl1 enhancer regions. Ankyloblepharon-ectodermal defects-cleft lip/palate (AEC) syndrome is a rare autosomal dominant disorder, caused by mutations in p63 gene, mainly characterized by skin fragility. To test whether nectins may be affected in AEC syndrome, their expression was measured in keratinocytes obtained from patients with AEC or from a conditional mouse model for AEC syndrome. Pvrl1 expression was reduced in AEC keratinocytes, consistent with impaired p63 function. Surprisingly, Pvrl4 expression was similarly affected, in parallel with decreased expression of the transcription factor Irf6. Consistent with the well-characterized role of Irf6 in keratinocyte differentiation and its strong downregulation in AEC syndrome, Irf6 depletion caused reduced expression of Pvrl4 in wild-type keratinocytes. Taken together, our results indicate that Pvrl1 is a bona fide target gene of the transcription factor p63, whereas Pvrl4 regulation is linked to epidermal differentiation and is under Irf6 control.


Subject(s)
Cell Adhesion Molecules/metabolism , Epidermis/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Animals, Newborn , Cells, Cultured , Epidermis/embryology , Gene Expression Regulation , Gene Expression Regulation, Developmental , Genes, Reporter , Humans , Keratinocytes/cytology , Mice , Mutation , Nectins , Phenotype , Phosphoproteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic
12.
Exp Dermatol ; 23(3): 143-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24417641

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC) is the second most common human cancer with a frequency increasing worldwide. The risk of developing cSCC has been strongly associated with chronic sun exposure, especially in light skin people. The aim of this viewpoint is to discuss the contribution of the tumor suppressor p53 and its homologues p63 and p73 in the formation and progression of cSCC. Mutations in the p53 gene are early and frequent events in skin carcinogenesis mainly as a consequence of UV light exposure, often followed by loss of function of the second allele. Although rarely mutated in cancer, p63 and p73 play key roles in human cancers, with their truncated isoforms lacking the N-terminal transactivating domain (∆N) being often upregulated as compared to normal tissues. ∆Np63 is abundantly expressed in cSCC, and it is likely to favour tumor initiation and progression. The function of p73 in cSCC is more enigmatic and awaits further studies. Interestingly, an intimate interplay exists between both p53 and p63, and the Notch signalling pathway, often inactivated in cSCC. Here, we summarize our current knowledge about the biological activities of p53 family members in cSCC and propose that integration of their signalling with Notch is key to cSCC formation and progression.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Squamous Cell/genetics , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Skin Neoplasms/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Animals , Carcinoma, Squamous Cell/metabolism , DNA-Binding Proteins/metabolism , Humans , Mutation , Nuclear Proteins/metabolism , Signal Transduction , Skin Neoplasms/metabolism , Transcription Factors/metabolism , Tumor Protein p73 , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
13.
Int Immunol ; 25(12): 703-14, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24038600

ABSTRACT

In humans, the thymus is the primary lymphoid organ able to support the development of T cells through its three-dimensional (3D) organization of the thymic stromal cells. Since a remarkable number of similarities are shared between the thymic epithelial cells (TECs) and skin-derived keratinocytes and fibroblasts, in this study we used human keratinocytes seeded with fibroblasts on the 3D poly ε-caprolactone scaffold to evaluate their ability to replace TECs in supporting T-cell differentiation from human haematopoietic stem cells (HSCs). We observed that in the multicellular biocomposite, early thymocytes expressing CD7(+)CD1a(+), peculiar markers of an initial T-cell commitment, were de novo generated. Molecular studies of genes selectively expressed during T-cell development revealed that TAL1 was down-regulated and Spi-B was up-regulated in the cell suspension, consistently with a T-cell lineage commitment. Moreover, PTCRA and RAG2 expression was detected, indicative of a recombinant activity, required for the generation of a T-cell receptor repertoire. Our results indicate that in the multicellular biocomposite, containing skin-derived elements in the absence of thymic stroma, HSCs do start differentiating toward a T-cell lineage commitment. In conclusion, the construct described in this study exerts some properties of a lymphoid organoid, suitable for future clinical applications in cell-based therapies.


Subject(s)
Caproates , Cell Differentiation , Fibroblasts/physiology , Hematopoietic Stem Cells/cytology , Keratinocytes/physiology , Lactones , Precursor Cells, T-Lymphoid/cytology , Tissue Scaffolds , Caproates/chemistry , Cell Communication , Cell Proliferation , Cell Survival , Coculture Techniques , Epidermal Cells , Fibroblasts/ultrastructure , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Keratinocytes/ultrastructure , Lactones/chemistry , Leukocytes, Mononuclear/physiology , Phenotype , Porosity , Precursor Cells, T-Lymphoid/metabolism , Thymus Gland/cytology , Thymus Gland/physiology , Tissue Scaffolds/chemistry
14.
Cell Death Dis ; 14(4): 274, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37072394

ABSTRACT

The transcription factor p63 shares a high sequence identity with the tumour suppressor p53 which manifests itself in high structural similarity and preference for DNA sequences. Mutations in the DNA binding domain (DBD) of p53 have been studied in great detail, enabling a general mechanism-based classification. In this study we provide a detailed investigation of all currently known mutations in the p63 DBD, which are associated with developmental syndromes, by measuring their impact on transcriptional activity, DNA binding affinity, zinc binding capacity and thermodynamic stability. Some of the mutations we have further characterized with respect to their ability to convert human dermal fibroblasts into induced keratinocytes. Here we propose a classification of the p63 DBD mutations based on the four different mechanisms of DNA binding impairment which we identified: direct DNA contact, zinc finger region, H2 region, and dimer interface mutations. The data also demonstrate that, in contrast to p53 cancer mutations, no p63 mutation induces global unfolding and subsequent aggregation of the domain. The dimer interface mutations that affect the DNA binding affinity by disturbing the interaction between the individual DBDs retain partial DNA binding capacity which correlates with a milder patient phenotype.


Subject(s)
Tumor Suppressor Protein p53 , Tumor Suppressor Proteins , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Protein Binding/genetics , Mutation/genetics , DNA/metabolism , Binding Sites
15.
Stem Cells ; 29(11): 1672-83, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21898690

ABSTRACT

p63, a member of the p53 family, is essential for skin morphogenesis and epithelial stem cell maintenance. Here, we report an unexpected role of TAp63 in cardiogenesis. p63 null mice exhibit severe defects in embryonic cardiac development, including dilation of both ventricles, a defect in trabeculation and abnormal septation. This was accompanied by myofibrillar disarray, mitochondrial disorganization, and reduction in spontaneous calcium spikes. By the use of embryonic stem cells (ESCs), we show that TAp63 deficiency prevents expression of pivotal cardiac genes and production of cardiomyocytes. TAp63 is expressed by endodermal cells. Coculture of p63-knockdown ESCs with wild-type ESCs, supplementation with Activin A, or overexpression of GATA-6 rescue cardiogenesis. Therefore, TAp63 acts in a non-cell-autonomous manner by modulating expression of endodermal factors. Our findings uncover a critical role for p63 in cardiogenesis that could be related to human heart disease.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Heart/embryology , Phosphoproteins/metabolism , Trans-Activators/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Embryonic Stem Cells/ultrastructure , Flow Cytometry , Fluorescent Antibody Technique , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , HMGB Proteins/genetics , HMGB Proteins/metabolism , Heart/growth & development , Immunoblotting , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Phosphoproteins/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Trans-Activators/genetics
16.
J Clin Invest ; 132(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-34905516

ABSTRACT

Desmoglein 1 (Dsg1) is a cadherin restricted to stratified tissues of terrestrial vertebrates, which serve as essential physical and immune barriers. Dsg1 loss-of-function mutations in humans result in skin lesions and multiple allergies, and isolated patient keratinocytes exhibit increased proallergic cytokine expression. However, the mechanism by which genetic deficiency of Dsg1 causes chronic inflammation is unknown. To determine the systemic response to Dsg1 loss, we deleted the 3 tandem Dsg1 genes in mice. Whole transcriptome analysis of embryonic Dsg1-/- skin showed a delay in expression of adhesion/differentiation/keratinization genes at E17.5, a subset of which recovered or increased by E18.5. Comparing epidermal transcriptomes from Dsg1-deficient mice and humans revealed a shared IL-17-skewed inflammatory signature. Although the impaired intercellular adhesion observed in Dsg1-/- mice resembles that resulting from anti-Dsg1 pemphigus foliaceus antibodies, pemphigus skin lesions exhibit a weaker IL-17 signature. Consistent with the clinical importance of these findings, treatment of 2 Dsg1-deficient patients with an IL-12/IL-23 antagonist originally developed for psoriasis resulted in improvement of skin lesions. Thus, beyond impairing the physical barrier, loss of Dsg1 function through gene mutation results in a psoriatic-like inflammatory signature before birth, and treatment with a targeted therapy significantly improved skin lesions in patients.


Subject(s)
Desmoglein 1/immunology , Desmosomes/immunology , Keratinocytes/immunology , Pemphigus/immunology , Th17 Cells/immunology , Animals , Desmoglein 1/genetics , Desmosomes/genetics , Mice , Pemphigus/genetics
18.
Cancers (Basel) ; 13(3)2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33572532

ABSTRACT

The p63 gene encodes a master regulator of epidermal commitment, development, and differentiation. Heterozygous mutations in the DNA binding domain cause Ectrodactyly, Ectodermal Dysplasia, characterized by limb deformation, cleft lip/palate, and ectodermal dysplasia while mutations in in the C-terminal domain of the α-isoform cause Ankyloblepharon-Ectodermal defects-Cleft lip/palate (AEC) syndrome, a life-threatening disorder characterized by skin fragility, severe, long-lasting skin erosions, and cleft lip/palate. The molecular disease mechanisms of these syndromes have recently become elucidated and have enhanced our understanding of the role of p63 in epidermal development. Here we review the molecular cause and functional consequences of these p63-mutations for skin development and discuss the consequences of p63 mutations for female fertility.

19.
Sci Rep ; 11(1): 2941, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536486

ABSTRACT

In recent months, Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread throughout the world. COVID-19 patients show mild, moderate or severe symptoms with the latter ones requiring access to specialized intensive care. SARS-CoV-2 infections, pathogenesis and progression have not been clearly elucidated yet, thus forcing the development of many complementary approaches to identify candidate cellular pathways involved in disease progression. Host lipids play a critical role in the virus life, being the double-membrane vesicles a key factor in coronavirus replication. Moreover, lipid biogenesis pathways affect receptor-mediated virus entry at the endosomal cell surface and modulate virus propagation. In this study, targeted lipidomic analysis coupled with proinflammatory cytokines and alarmins measurement were carried out in serum of COVID-19 patients characterized by different severity degree. Serum IL-26, a cytokine involved in IL-17 pathway, TSLP and adiponectin were measured and correlated to lipid COVID-19 patient profiles. These results could be important for the classification of the COVID-19 disease and the identification of therapeutic targets.


Subject(s)
COVID-19/pathology , Lipid Metabolism/physiology , Alarmins/blood , COVID-19/virology , Cytokines/blood , Discriminant Analysis , Female , Humans , Least-Squares Analysis , Lipids/blood , Male , Middle Aged , SARS-CoV-2/isolation & purification , Severity of Illness Index
20.
J Biol Chem ; 284(44): 30574-82, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19717565

ABSTRACT

p63, a p53 family member, plays an essential role in epidermal development by regulating its transcriptional program. Here we report a previously uncovered role of p63 in controlling bone morphogenetic protein (BMP) signaling, which is required for maintaining low expression levels of several non-epidermal genes. p63 represses transcription of the inhibitory Smad7 and activates Bmp7, thereby sustaining BMP signaling. In the absence of p63, compromised BMP signaling leads to inappropriate non-epidermal gene expression in postnatal mouse keratinocytes and in embryonic epidermis. Reactivation of BMP signaling by Smad7 knockdown and/or, to a lesser extent, by BMP treatment suppresses expression of non-epidermal genes in the absence of p63. Canonical BMP/Smad signaling is essential for control of non-epidermal genes as use of a specific inhibitor, or simultaneous knockdown of Smad1 and Smad5 counteract suppression of non-epidermal genes. Our data indicate that p63 prevents ectopic expression of non-epidermal genes by a mechanism involving Smad7 repression and, to a lesser extent, Bmp7 induction, with consequent enhancement of BMP/Smad signaling.


Subject(s)
Bone Morphogenetic Protein 7/physiology , Epidermal Cells , Phosphoproteins/physiology , Smad7 Protein/antagonists & inhibitors , Trans-Activators/physiology , Animals , Biomarkers , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Proteins/physiology , Cell Lineage , Cells, Cultured , Gene Expression Regulation , Keratinocytes/cytology , Mice , Signal Transduction , Smad7 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL