Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Immunity ; 45(1): 119-30, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27438769

RESUMO

Type 1 interferons (IFNs) promote inflammation in the skin but the mechanisms responsible for inducing these cytokines are not well understood. We found that IFN-ß was abundantly produced by epidermal keratinocytes (KCs) in psoriasis and during wound repair. KC IFN-ß production depended on stimulation of mitochondrial antiviral-signaling protein (MAVS) by the antimicrobial peptide LL37 and double stranded-RNA released from necrotic cells. MAVS activated downstream TBK1 (TANK-Binding Kinase 1)-AKT (AKT serine/threonine kinase 1)-IRF3 (interferon regulatory factor 3) signaling cascade leading to IFN-ß production and then promoted maturation of dendritic cells. In mice, the production of epidermal IFN-ß by LL37 required MAVS, and human wounded and/or psoriatic skin showed activation of MAVS-associated IRF3 and induction of MAVS and IFN-ß gene signatures. These findings show that KCs are an important source of IFN-ß and MAVS is critical to this function, and demonstrates how the epidermis triggers unwanted skin inflammation under disease conditions.


Assuntos
Catelicidinas/metabolismo , Células Dendríticas/fisiologia , Epiderme/patologia , Queratinócitos/imunologia , Mitocôndrias/metabolismo , Psoríase/imunologia , Ferimentos e Lesões/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos , Catelicidinas/genética , Diferenciação Celular , Células Cultivadas , Humanos , Interferon beta/metabolismo , Camundongos , Camundongos Knockout , RNA Interferente Pequeno/genética , Transdução de Sinais , Cicatrização
2.
PLoS Biol ; 19(9): e3001378, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34543262

RESUMO

Stratified epithelia such as the epidermis require coordinated regulation of stem and progenitor cell proliferation, survival, and differentiation to maintain homeostasis. Integrin-mediated anchorage of the basal layer stem cells of the epidermis to the underlying dermis through extracellular matrix (ECM) proteins is crucial for this process. It is currently unknown how the expression of these integrins and ECM genes are regulated. Here, we show that the RNA-binding protein (RBP) heterogeneous nuclear ribonucleoprotein L (HNRNPL) binds to these genes on chromatin to promote their expression. HNRNPL recruits RNA polymerase II (Pol II) to integrin/ECM genes and is required for stabilizing Pol II transcription through those genes. In the absence of HNRNPL, the basal layer of the epidermis where the stem cells reside prematurely differentiates and detaches from the underlying dermis due to diminished integrin/ECM expression. Our results demonstrate a critical role for RBPs on chromatin to maintain stem and progenitor cell fate by dictating the expression of specific classes of genes.


Assuntos
Células Epidérmicas/metabolismo , Matriz Extracelular/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Integrinas/metabolismo , Diferenciação Celular , Células Cultivadas , Cromatina , Epiderme/crescimento & desenvolvimento , Matriz Extracelular/genética , Humanos , Integrinas/genética , Células-Tronco
3.
Stem Cells ; 40(4): 435-445, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-35325240

RESUMO

Proper differentiation of the epidermis is essential to prevent water loss and to protect the body from the outside environment. Perturbations in this process can lead to a variety of skin diseases that impacts 1 in 5 people. While transcription factors that control epidermal differentiation have been well characterized, other aspects of transcription control such as elongation are poorly understood. Here we show that of the two cyclin-dependent kinases (CDK12 and CDK13), that are known to regulate transcription elongation, only CDK12 is necessary for epidermal differentiation. Depletion of CDK12 led to loss of differentiation gene expression and absence of skin barrier formation in regenerated human epidermis. CDK12 binds to genes that code for differentiation promoting transcription factors (GRHL3, KLF4, and OVOL1) and is necessary for their elongation. CDK12 is necessary for elongation by promoting Ser2 phosphorylation on the C-terminal domain of RNA polymerase II and the stabilization of binding of the elongation factor SPT6 to target genes. Our results suggest that control of transcription elongation by CDK12 plays a prominent role in adult cell fate decisions.


Assuntos
Quinases Ciclina-Dependentes , RNA Polimerase II , Diferenciação Celular/genética , Quinases Ciclina-Dependentes/genética , Quinases Ciclina-Dependentes/metabolismo , Humanos , Fosforilação , RNA Polimerase II/química , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Mol Cell ; 60(1): 118-30, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26412305

RESUMO

In adult tissues, stem and progenitor cells must balance proliferation and differentiation to maintain homeostasis. How this is done is unclear. Here, we show that the DEAD box RNA helicase, DDX6 is necessary for maintaining adult progenitor cell function. DDX6 loss results in premature differentiation and decreased proliferation of epidermal progenitor cells. To maintain self-renewal, DDX6 associates with YBX1 to bind the stem loops found in the 3' UTRs of regulators of proliferation/self-renewal (CDK1, EZH2) and recruit them to EIF4E to facilitate their translation. To prevent premature differentiation of progenitor cells, DDX6 regulates the 5' UTR of differentiation inducing transcription factor, KLF4 and degrades its transcripts through association with mRNA degradation proteins. Our results demonstrate that progenitor function is maintained by DDX6 complexes through two distinct pathways that include the degradation of differentiation-inducing transcripts and by promoting the translation of self-renewal and proliferation mRNAs.


Assuntos
RNA Helicases DEAD-box/metabolismo , Epiderme/fisiologia , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas/metabolismo , Estabilidade de RNA , RNA Mensageiro/metabolismo , Células-Tronco/metabolismo , Autorrenovação Celular , Células Cultivadas , Fator de Iniciação 4E em Eucariotos/metabolismo , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Polirribossomos/metabolismo , Dobramento de RNA , RNA Mensageiro/química , Proteína 1 de Ligação a Y-Box/metabolismo
5.
J Cell Sci ; 133(23)2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-33172988

RESUMO

Proper epithelial development and homeostasis depends on strict control of oriented cell division. Current evidence shows that this process is regulated by intrinsic polarity factors and external spatial cues. Owing to the lack of an appropriate model system that can recapitulate the architecture of the skin, deregulation of spindle orientation in human epithelial carcinoma has never been investigated. Here, using an inducible model of human squamous cell carcinoma (SCC), we demonstrate that RAS-dependent suppression of PAR3 (encoded by PARD3) accelerates epithelial disorganization during early tumorigenesis. Diminished PAR3 led to loss of E-cadherin-mediated cell adhesion, which in turn contributed to misoriented cell division. Pharmacological inhibition of the MAPK pathway downstream of RAS activation reversed the defects in PAR3 expression, E-cadherin-mediated cell adhesion and mitotic spindle orientation. Thus, temporal analysis of human neoplasia provides a powerful approach to study cellular and molecular transformations during early oncogenesis, which allowed identification of PAR3 as a critical regulator of tissue architecture during initial human SCC development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Carcinoma de Células Escamosas , Proteínas de Ciclo Celular , Proteínas ras , Carcinogênese/genética , Carcinoma de Células Escamosas/genética , Adesão Celular , Proteínas de Ciclo Celular/metabolismo , Divisão Celular , Polaridade Celular , Humanos , Hiperplasia , Fuso Acromático/metabolismo
6.
Exp Dermatol ; 26(9): 792-797, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28094886

RESUMO

Darier disease (DD) is a genetic skin disease that is associated with mutations in the ATP2A2 gene encoding the type 2 sarco/endoplasmic reticulum (ER) Ca2+ - ATPase (SERCA2). Mutations of this gene result in alterations of calcium homoeostasis, abnormal epidermal adhesion and dyskeratosis. Silencing of ATP2A2 in monolayer cell culture of keratinocytes reduces desmoplakin expression at the borders of cells and impacts cell adhesion. Here, we report establishment of a three-dimensional (3D) epidermal model of DD and use this model to evaluate peptide therapy with tuberoinfundibular peptide of 39 residues (TIP39) to normalize calcium transport. Gene silencing of ATP2A2 in keratinocytes grown in a 3D model resulted in dyskeratosis, partial parakeratosis and suprabasal clefts that resembled the histological changes seen in skin biopsies from patients with DD. TIP39, a peptide recently identified as a regulator of keratinocyte calcium transport, was then applied to this ATP2A2-silenced 3D epidermal model. In normal keratinocytes, TIP39 increased [Ca2+ ]i through the inositol trisphosphate (IP3) receptor pathway and stimulated differentiation. In monolayer ATP2A2-silenced keratinocytes, although TIP39 increased cytosolic calcium from the ER, the response was incomplete compared with its control. TIP39 was observed to reduce intercellular clefts of the gene-silenced epidermal model but did not significantly upregulate keratinocyte differentiation genes such as keratin 10 and filaggrin. These findings indicate that TIP39 is a modulator of ER calcium signalling and may be used as a potential strategy for improving aspects of DD.


Assuntos
Cálcio/metabolismo , Doença de Darier/metabolismo , Neuropeptídeos/metabolismo , Receptor Tipo 2 de Hormônio Paratireóideo/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Células Cultivadas , Retículo Endoplasmático/metabolismo , Epiderme/metabolismo , Proteínas Filagrinas , Humanos , Queratinócitos/metabolismo
7.
J Biol Chem ; 290(33): 20448-54, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26045558

RESUMO

PAX6 is a master regulatory gene involved in neuronal cell fate specification. It also plays a critical role in early eye field and subsequent limbal stem cell (LSC) determination during eye development. Defects in Pax6 cause aniridia and LSC deficiency in humans and the Sey (Small eye) phenotype in mice (Massé, K., Bhamra, S., Eason, R., Dale, N., and Jones, E. A. (2007) Nature 449, 1058-1062). However, how PAX6 specifies LSC and corneal fates during eye development is not well understood. Here, we show that PAX6 is expressed in the primitive eye cup and later in corneal tissue progenitors in early embryonic development. In contrast, p63 expression commences after that of PAX6 in ocular adnexal and skin tissue progenitors and later in LSCs. Using an in vitro feeder-free culture system, we show that PAX6 knockdown in LSCs led to up-regulation of skin epidermis-specific keratins concomitant with differentiation to a skin fate. Using gene expression analysis, we identified the involvement of Notch, Wnt, and TGF-ß signaling pathways in LSC fate determination. Thus, loss of PAX6 converts LSCs to epidermal stem cells, as demonstrated by a switch in the keratin gene expression profile and by the appearance of congenital dermoid tissue.


Assuntos
Linhagem da Célula/fisiologia , Proteínas do Olho/fisiologia , Proteínas de Homeodomínio/fisiologia , Sistema Límbico/citologia , Fatores de Transcrição Box Pareados/fisiologia , Proteínas Repressoras/fisiologia , Células-Tronco/citologia , Animais , Córnea/embriologia , Proteínas do Olho/genética , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Sistema Límbico/metabolismo , Proteínas de Membrana/genética , Camundongos , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Proteínas Repressoras/genética , Transdução de Sinais , Células-Tronco/metabolismo
8.
Nature ; 463(7280): 563-7, 2010 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-20081831

RESUMO

Progenitor cells maintain self-renewing tissues throughout life by sustaining their capacity for proliferation while suppressing cell cycle exit and terminal differentiation. DNA methylation provides a potential epigenetic mechanism for the cellular memory needed to preserve the somatic progenitor state through repeated cell divisions. DNA methyltransferase 1 (DNMT1) maintains DNA methylation patterns after cellular replication. Although dispensable for embryonic stem cell maintenance, the role for DNMT1 in maintaining the progenitor state in constantly replenished somatic tissues, such as mammalian epidermis, is unclear. Here we show that DNMT1 is essential for epidermal progenitor cell function. DNMT1 protein was found enriched in undifferentiated cells, where it was required to retain proliferative stamina and suppress differentiation. In tissue, DNMT1 depletion led to exit from the progenitor cell compartment, premature differentiation and eventual tissue loss. Genome-wide analysis showed that a significant portion of epidermal differentiation gene promoters were methylated in self-renewing conditions but were subsequently demethylated during differentiation. Furthermore, UHRF1 (refs 9, 10), a component of the DNA methylation machinery that targets DNMT1 to hemi-methylated DNA, is also necessary to suppress premature differentiation and sustain proliferation. In contrast, Gadd45A and B, which promote active DNA demethylation, are required for full epidermal differentiation gene induction. These data demonstrate that proteins involved in the dynamic regulation of DNA methylation patterns are required for progenitor maintenance and self-renewal in mammalian somatic tissue.


Assuntos
Diferenciação Celular , Células Epidérmicas , Epiderme/metabolismo , Proteínas Repressoras/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Metilação de DNA , Regulação para Baixo , Feminino , Inativação Gênica , Humanos , Camundongos , Camundongos SCID , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética
9.
Am J Hum Genet ; 91(3): 435-43, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22922031

RESUMO

The basis for impaired differentiation in TP63 mutant ankyloblepharon-ectodermal dysplasia-clefting (AEC) syndrome is unknown. Human epidermis harboring AEC TP63 mutants recapitulated this impairment, along with downregulation of differentiation activators, including HOPX, GRHL3, KLF4, PRDM1, and ZNF750. Gene-set enrichment analysis indicated that disrupted expression of epidermal differentiation programs under the control of ZNF750 and KLF4 accounted for the majority of disrupted epidermal differentiation resulting from AEC mutant TP63. Chromatin immunoprecipitation (ChIP) analysis and ChIP-sequencing of TP63 binding in differentiated keratinocytes revealed ZNF750 as a direct target of wild-type and AEC mutant TP63. Restoring ZNF750 to AEC model tissue rescued activator expression and differentiation, indicating that AEC TP63-mediated ZNF750 inhibition contributes to differentiation defects in AEC. Incorporating disease-causing mutants into regenerated human tissue can thus dissect pathomechanisms and identify targets that reverse disease features.


Assuntos
Fenda Labial/genética , Fissura Palatina/genética , Displasia Ectodérmica/genética , Anormalidades do Olho/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Diferenciação Celular/genética , Epiderme/metabolismo , Pálpebras/anormalidades , Humanos , Fator 4 Semelhante a Kruppel , Mutação , Técnicas de Cultura de Órgãos/métodos , Transcriptoma
10.
Stem Cells ; 32(12): 3209-18, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25100569

RESUMO

The transcription factor, SNAI2, is an inducer of the epithelial to mesenchymal transition (EMT) which mediates cell migration during development and tumor invasion. SNAI2 can also promote the generation of mammary epithelial stem cells from differentiated luminal cells when overexpressed. How SNAI2 regulates these critical and diverse functions is unclear. Here, we show that the levels of SNAI2 expression are important for epidermal cell fate decisions. The expression of SNAI2 was found to be enriched in the basal layer of the interfollicular epidermis where progenitor cells reside and extinguished upon differentiation. Loss of SNAI2 resulted in premature differentiation whereas gain of SNAI2 expression inhibited differentiation. SNAI2 controls the differentiation status of epidermal progenitor cells by binding to and repressing the expression of differentiation genes with increased binding leading to further transcriptional silencing. Thus, the levels of SNAI2 binding to genomic targets determine the differentiation status of epithelial cells with increased levels triggering EMT and dedifferentiation, moderate (physiological) levels promoting epidermal progenitor function, and low levels leading to epidermal differentiation.


Assuntos
Diferenciação Celular/fisiologia , Células Epiteliais/citologia , Transição Epitelial-Mesenquimal/fisiologia , Células-Tronco/citologia , Fatores de Transcrição/metabolismo , Linhagem Celular , Movimento Celular/fisiologia , Células Epiteliais/metabolismo , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição da Família Snail , Células-Tronco/metabolismo
11.
Cell Rep ; 42(5): 112511, 2023 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-37195865

RESUMO

Several methods for generating human-skin-equivalent (HSE) organoid cultures are in use to study skin biology; however, few studies thoroughly characterize these systems. To fill this gap, we use single-cell transcriptomics to compare in vitro HSEs, xenograft HSEs, and in vivo epidermis. By combining differential gene expression, pseudotime analyses, and spatial localization, we reconstruct HSE keratinocyte differentiation trajectories that recapitulate known in vivo epidermal differentiation pathways and show that HSEs contain major in vivo cellular states. However, HSEs also develop unique keratinocyte states, an expanded basal stem cell program, and disrupted terminal differentiation. Cell-cell communication modeling shows aberrant epithelial-to-mesenchymal transition (EMT)-associated signaling pathways that alter upon epidermal growth factor (EGF) supplementation. Last, xenograft HSEs at early time points post transplantation significantly rescue many in vitro deficits while undergoing a hypoxic response that drives an alternative differentiation lineage. This study highlights the strengths and limitations of organoid cultures and identifies areas for potential innovation.


Assuntos
Pele , Transcriptoma , Humanos , Transcriptoma/genética , Pele/metabolismo , Queratinócitos/metabolismo , Epiderme/metabolismo , Diferenciação Celular , Organoides
12.
Nat Cell Biol ; 7(6): 633-6, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15908945

RESUMO

RNA interference (RNAi) is an important means of eliminating mRNAs, but the intracellular location of RNA-induced silencing complex (RISC) remains unknown. We show here that Argonaute 2, a key component of RISC, is not randomly distributed but concentrates in mRNA decay centres that are known as cytoplasmic bodies. The localization of Argonaute 2 in decay centres is not altered by the presence or absence of small interfering RNAs or their targeted mRNAs. However, RNA is required for the integrity of cytoplasmic bodies because RNase eliminates Argonaute 2 localization. In addition, Argonaute 1, another Argonaute family member, is concentrated in cytoplasmic bodies. These results provide new insight into the mechanism of RNAi function.


Assuntos
Citoplasma/metabolismo , Fatores de Iniciação de Peptídeos/metabolismo , Interferência de RNA/fisiologia , RNA Mensageiro/metabolismo , Complexo de Inativação Induzido por RNA/metabolismo , Animais , Proteínas Argonautas , Autoantígenos/genética , Autoantígenos/metabolismo , Citoplasma/genética , Fator de Iniciação 2 em Eucariotos , Humanos , Substâncias Macromoleculares/metabolismo , Camundongos , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Fatores de Iniciação de Peptídeos/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA , Complexo de Inativação Induzido por RNA/genética
13.
FASEB J ; 25(7): 2123-8, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21441350

RESUMO

Stem cells serve a vital role in multicellular organisms by constantly replenishing tissue with their differentiated progeny during normal homeostasis or damage. How these cells maintain their identities throughout the life of an organism is an area of intense research. In this review, we explore recent emerging evidence that stem cell fate decisions are based on their epigenome and specific epigenetic factors.


Assuntos
Diferenciação Celular , Proliferação de Células , Células-Tronco/citologia , Células-Tronco/metabolismo , Metilação de DNA , DNA-Citosina Metilases/metabolismo , Histona Desmetilases/metabolismo , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Humanos , Metilação
14.
Nat Commun ; 12(1): 784, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33542242

RESUMO

In adult tissue, stem and progenitor cells must tightly regulate the balance between proliferation and differentiation to sustain homeostasis. How this exquisite balance is achieved is an area of active investigation. Here, we show that epidermal genes, including ~30% of induced differentiation genes already contain stalled Pol II at the promoters in epidermal stem and progenitor cells which is then released into productive transcription elongation upon differentiation. Central to this process are SPT6 and PAF1 which are necessary for the elongation of these differentiation genes. Upon SPT6 or PAF1 depletion there is a loss of human skin differentiation and stratification. Unexpectedly, loss of SPT6 also causes the spontaneous transdifferentiation of epidermal cells into an intestinal-like phenotype due to the stalled transcription of the master regulator of epidermal fate P63. Our findings suggest that control of transcription elongation through SPT6 plays a prominent role in adult somatic tissue differentiation and the inhibition of alternative cell fate choices.


Assuntos
Diferenciação Celular/genética , Epiderme/fisiologia , Elongação da Transcrição Genética , Fatores de Transcrição/metabolismo , Células-Tronco Adultas/fisiologia , Transdiferenciação Celular/genética , Células Cultivadas , Sequenciamento de Cromatina por Imunoprecipitação , Técnicas de Silenciamento de Genes , Humanos , Recém-Nascido , Queratinócitos , Masculino , Cultura Primária de Células , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , RNA Interferente Pequeno/metabolismo , RNA-Seq , Técnicas de Cultura de Tecidos , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/metabolismo
16.
iScience ; 23(7): 101320, 2020 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-32659720

RESUMO

Impairments in the differentiation process can lead to skin diseases that can afflict ∼20% of the population. Thus, it is of utmost importance to understand the factors that promote the differentiation process. Here we identify the transcription factor KLF3 as a regulator of epidermal differentiation. Knockdown of KLF3 results in reduced differentiation gene expression and increased cell cycle gene expression. Over half of KLF3's genomic binding sites occur at active enhancers. KLF3 binds to active enhancers proximal to differentiation genes that are dependent upon KLF3 for expression. KLF3's genomic binding sites also highly overlaps with CBP, a histone acetyltransferase necessary for activating enhancers. Depletion of KLF3 causes reduced CBP localization at enhancers proximal to differentiation gene clusters, which leads to loss of enhancer activation but not priming. Our results suggest that KLF3 is necessary to recruit CBP to activate enhancers and drive epidermal differentiation gene expression.

17.
Nat Commun ; 10(1): 4198, 2019 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-31519929

RESUMO

Maintenance of high-turnover tissues such as the epidermis requires a balance between stem cell proliferation and differentiation. The molecular mechanisms governing this process are an area of investigation. Here we show that HNRNPK, a multifunctional protein, is necessary to prevent premature differentiation and sustains the proliferative capacity of epidermal stem and progenitor cells. To prevent premature differentiation of progenitor cells, HNRNPK is necessary for DDX6 to bind a subset of mRNAs that code for transcription factors that promote differentiation. Upon binding, these mRNAs such as GRHL3, KLF4, and ZNF750 are degraded through the mRNA degradation pathway, which prevents premature differentiation. To sustain the proliferative capacity of the epidermis, HNRNPK is necessary for RNA Polymerase II binding to proliferation/self-renewal genes such as MYC, CYR61, FGFBP1, EGFR, and cyclins to promote their expression. Our study establishes a prominent role for HNRNPK in maintaining adult tissue self-renewal through both transcriptional and post-transcriptional mechanisms.


Assuntos
Diferenciação Celular/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , RNA Mensageiro/metabolismo , Diferenciação Celular/genética , Proliferação de Células/genética , Proliferação de Células/fisiologia , Células Cultivadas , Proteína Rica em Cisteína 61/genética , Proteína Rica em Cisteína 61/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Epiderme/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/genética , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Estabilidade de RNA/genética , Estabilidade de RNA/fisiologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor
19.
ACS Nano ; 13(6): 6605-6617, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31188564

RESUMO

Stem cell therapy in heart disease is challenged by mis-injection, poor survival, and low cell retention. Here, we describe a biocompatible multifunctional silica-iron oxide nanoparticle to help solve these issues. The nanoparticles were made via an in situ growth of Fe3O4 nanoparticles on both the external surfaces and pore walls of mesocellular foam silica nanoparticles. In contrast to previous work, this approach builds a magnetic moiety inside the pores of a porous silica structure. These materials serve three roles: drug delivery, magnetic manipulation, and imaging. The addition of Fe3O4 to the silica nanoparticles increased their colloidal stability, T2-based magnetic resonance imaging contrast, and superparamagnetism. We then used the hybrid materials as a sustained release vehicle of insulin-like growth factor-a pro-survival agent that can increase cell viability. In vivo rodent studies show that labeling stem cells with this nanoparticle increased the efficacy of stem cell therapy in a ligation/reperfusion model. The nanoparticle-labeled cells increase the mean left ventricular ejection fraction by 11 and 21% and the global longitudinal strain by 24 and 34% on days 30 and 60, respectively. In summary, this multifunctional nanomedicine improves stem cell survival via the sustained release of pro-survival agents.


Assuntos
Fator de Crescimento Insulin-Like I/administração & dosagem , Transplante de Células-Tronco Mesenquimais/métodos , Nanopartículas/química , Nanomedicina Teranóstica/métodos , Animais , Células Cultivadas , Meios de Contraste/química , Liberação Controlada de Fármacos , Compostos Férricos/química , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Imageamento por Ressonância Magnética/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/terapia , Dióxido de Silício/química
20.
Biomaterials ; 179: 60-70, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29980075

RESUMO

Silicon carbide has been shown to be biocompatible and is used as a coating material for implanted medical devices to prevent biofilms. Silicon carbide nanomaterials are also promising in cell tracking due to their stable and strong luminescence, but more comprehensive studies of this material on the nanoscale are needed. Here, we studied the toxicity of silicon carbide nanomaterials on human mesenchymal stem cells in terms of metabolism, viability, adhesion, proliferation, migration, oxidative stress, and differentiation ability. We compared two different shapes and found that silicon carbide nanowires are toxic to human mesenchymal stem cells but not to cancer cell lines at the concentration of 0.1 mg/mL. Control silicon carbide nanoparticles were biocompatible to human mesenchymal stem cells at 0.1 mg/mL. We studied the potential mechanistic effect of silicon carbide nanowires on human mesenchymal stem cells' phenotype, cytokine secretion, and gene expression. These findings suggest that the toxic effect of silicon carbide nanomaterials to human mesenchymal stem cells are dependent on morphology.


Assuntos
Compostos Inorgânicos de Carbono/química , Nanoestruturas/química , Nanofios/química , Compostos de Silício/química , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Citocinas/química , Humanos , Células-Tronco Mesenquimais/citologia , Estresse Oxidativo/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA