Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 27(10): 1706-1717, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31526597

RESUMO

The field of cell and gene therapy (GT) is expanding rapidly and there is undoubtedly a wave of enthusiasm and anticipation for what these treatments could achieve next. Here we assessed the worldwide landscape of GT assets currently in early clinical development (clinical trial phase 1/2 or about to enter clinical trial). We included all gene therapies, i.e., strategies that modify an individual's protein make-up by introducing exogenous nucleic acid or nucleic acid modifiers, regardless of delivery. Unmodified cell therapies, oncology therapies (reviewed elsewhere), and vaccine programs (distinct therapeutic strategy) were not included. Using a December 31, 2018 cutoff date, we identified 336 gene therapies being developed for 138 different indications covering 165 genetic targets. In all, we found that the early clinical GT landscape comprises a very disparate group of drug candidates in terms of indications, organizations, and delivery methods. We also highlight interesting trends, revealing the evolution of the field toward in vivo therapies and adeno-associated virus vector-based delivery systems. It will be interesting to witness what proportion of this current list effectively translates into new medicines.


Assuntos
Sistemas de Liberação de Medicamentos/classificação , Terapia Genética/métodos , Ensaios Clínicos como Assunto , Vetores Genéticos/administração & dosagem , Humanos , Terapia de Alvo Molecular
2.
Exp Dermatol ; 25(3): 187-93, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26519132

RESUMO

To explore the role of amphiregulin in inflammatory epidermal hyperplasia, we overexpressed human AREG (hAREG) in FVB/N mice using a bovine K5 promoter. A construct containing AREG coding sequences flanked by 5' and 3' untranslated region sequences (AREG-UTR) led to a >10-fold increase in hAREG expression compared to an otherwise-identical construct containing only the coding region (AREG-CDR). AREG-UTR mice developed tousled, greasy fur as well as elongated nails and thickened, erythematous tail skin. No such phenotype was evident in AREG-CDR mice. Histologically, AREG-UTR mice presented with marked epidermal hyperplasia of tail skin (2.1-fold increase in epidermal thickness with a 9.5-fold increase in Ki-67(+) cells) accompanied by significantly increased CD4+ T-cell infiltration. Dorsal skin of AREG-UTR mice manifested lesser but still significant increases in epidermal thickness and keratinocyte hyperplasia. AREG-UTR mice also developed marked and significant sebaceous gland enlargement, with corresponding increases in Ki-67(+) cells. To determine the response of AREG-UTR animals to a pro-inflammatory skin challenge, topical imiquimod (IMQ) or vehicle cream was applied to dorsal and tail skin. IMQ increased dorsal skin thickness similarly in both AREG-UTR and wild type mice (1.7- and 2.2-fold vs vehicle, P < 0.001 each), but had no such effect on tail skin. These results confirm that keratinocyte expression of hAREG elicits inflammatory epidermal hyperplasia, and are consistent with prior reports of tail epidermal hyperplasia and increased sebaceous gland size in mice expressing human epigen.


Assuntos
Anfirregulina/genética , Anfirregulina/metabolismo , Pele/metabolismo , Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Animais , Bovinos , Epiderme/patologia , Receptores ErbB/metabolismo , Homeostase , Humanos , Hiperplasia/metabolismo , Inflamação , Ligantes , Camundongos , Camundongos Transgênicos , Fenótipo , Regiões Promotoras Genéticas , Glândulas Sebáceas/metabolismo
3.
Am J Pathol ; 182(1): 163-71, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23159944

RESUMO

Eccrine sweat glands are skin-associated epithelial structures (appendages) that are unique to some primates including humans and are absent in the skin of most laboratory animals including rodents, rabbits, and pigs. On the basis of the known importance of other skin appendages (hair follicles, apocrine glands, and sebaceous glands) for wound repair in model animals, the present study was designed to assess the role of eccrine glands in the repair of wounded human skin. Partial-thickness wounds were generated on healthy human forearms, and epidermal repair was studied in skin biopsy samples obtained at precise times during the first week after wounding. Wound reepithelialization was assessed using immunohistochemistry and computer-assisted 3-dimensional reconstruction of in vivo wounded skin samples. Our data demonstrate a key role for eccrine sweat glands in reconstituting the epidermis after wounding in humans. More specifically, (i) eccrine sweat glands generate keratinocyte outgrowths that ultimately form new epidermis; (ii) eccrine sweat glands are the most abundant appendages in human skin, outnumbering hair follicles by a factor close to 3; and (iii) the rate of expansion of keratinocyte outgrowths from eccrine sweat glands parallels the rate of reepithelialization. This novel appreciation of the unique importance of eccrine sweat glands for epidermal repair may be exploited to improve our approaches to understanding and treating human wounds.


Assuntos
Glândulas Écrinas/fisiologia , Reepitelização/fisiologia , Pele/lesões , Adolescente , Adulto , Biópsia , Proliferação de Células , Glândulas Écrinas/patologia , Epiderme/patologia , Epiderme/fisiologia , Feminino , Humanos , Queratinócitos/patologia , Masculino , Pessoa de Meia-Idade , Pele/patologia , Adulto Jovem
4.
JCI Insight ; 8(14)2023 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-37485877

RESUMO

Keratin (K) and other intermediate filament (IF) protein mutations at conserved arginines disrupt keratin filaments into aggregates and cause human epidermolysis bullosa simplex (EBS; K14-R125C) or predispose to mouse liver injury (K18-R90C). The challenge for more than 70 IF-associated diseases is the lack of clinically utilized IF-targeted therapies. We used high-throughput drug screening to identify compounds that normalized mutation-triggered keratin filament disruption. Parthenolide, a plant sesquiterpene lactone, dramatically reversed keratin filament disruption and protected cells and mice expressing K18-R90C from apoptosis. K18-R90C became hyperacetylated compared with K18-WT and treatment with parthenolide normalized K18 acetylation. Parthenolide upregulated the NAD-dependent SIRT2, and increased SIRT2-keratin association. SIRT2 knockdown or pharmacologic inhibition blocked the parthenolide effect, while site-specific Lys-to-Arg mutation of keratin acetylation sites normalized K18-R90C filaments. Treatment of K18-R90C-expressing cells and mice with nicotinamide mononucleotide had a parthenolide-like protective effect. In 2 human K18 variants that associate with human fatal drug-induced liver injury, parthenolide protected K18-D89H- but not K8-K393R-induced filament disruption and cell death. Importantly, parthenolide normalized K14-R125C-mediated filament disruption in keratinocytes and inhibited dispase-triggered keratinocyte sheet fragmentation and Fas-mediated apoptosis. Therefore, keratin acetylation may provide a novel therapeutic target for some keratin-associated diseases.


Assuntos
Queratinas , Sirtuína 2 , Animais , Humanos , Camundongos , Proteínas de Filamentos Intermediários , Queratinas/genética , Queratinas/metabolismo , Mutação , Sirtuína 2/genética
5.
Arthritis Rheum ; 63(10): 3022-31, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21618206

RESUMO

OBJECTIVE: To investigate transforming growth factor ß (TGFß) regulation of CCN3 expression in cells of the nucleus pulposus. METHODS: Real-time reverse transcription-polymerase chain reaction and Western blot analyses were used to measure CCN3 expression in the nucleus pulposus. Transfections were used to measure the effect of Smad3, MAPKs, and activator protein 1 (AP-1) on TGFß-mediated CCN3 promoter activity. Lentiviral knockdown of Smad3 was performed to assess the role of Smad3 in CCN3 expression. RESULTS: CCN3 was expressed in embryonic and adult intervertebral discs. TGFß decreased the expression of CCN3 and suppressed its promoter activity in nucleus pulposus cells. DN-Smad3, Smad3 small interfering RNA, or DN-AP-1 had little effect on TGFß suppression of CCN3 promoter activity. However, p38 and ERK inhibitors blocked suppression of CCN3 by TGFß, suggesting involvement of these signaling pathways in the regulation of CCN3. Interestingly, overexpression of Smad3 in the absence of TGFß increased CCN3 promoter activity. We validated the role of Smad3 in controlling CCN3 expression in Smad3-null mice and in nucleus pulposus cells transduced with lentiviral short hairpin Smad3. In terms of function, treatment with recombinant CCN3 showed a dose-dependent decrease in the proliferation of nucleus pulposus cells. Moreover, CCN3-treated cells showed a decrease in aggrecan, versican, CCN2, and type I collagen expression. CONCLUSION: The opposing effect of TGFß on CCN2 and CCN3 expression and the suppression of CCN2 by CCN3 in nucleus pulposus cells further the paradigm that these CCN proteins form an interacting triad, which is possibly important in maintaining extracellular matrix homeostasis and cell numbers.


Assuntos
Disco Intervertebral/metabolismo , Proteína Sobre-Expressa em Nefroblastoma/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Disco Intervertebral/citologia , Disco Intervertebral/efeitos dos fármacos , Camundongos , Proteína Sobre-Expressa em Nefroblastoma/genética , Proteína Sobre-Expressa em Nefroblastoma/farmacologia , Regiões Promotoras Genéticas , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Fator de Crescimento Transformador beta/genética
6.
J Am Acad Dermatol ; 64(1): 119-28, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21167407

RESUMO

BACKGROUND: Various minimally invasive treatments enhance the skin's appearance. Little is known about the molecular mechanisms whereby treatments working at the epidermal level might alter the dermis. OBJECTIVE: We sought to quantify the molecular changes that result from erbium:yttrium-aluminium-garnet (Er:YAG) laser microablative resurfacing. METHODS: We performed biochemical analyses after intraepidermal Er:YAG laser resurfacing of 10 patients. Immunohistochemical analysis and polymerase chain reaction technology were utilized to measure key biomarkers. RESULTS: The basement membrane remained intact after intraepidermal microablation, as demonstrated by laminin γ2 immunostaining. Epidermal injury was demonstrated with acute up-regulation of keratin 16. An inflammatory response ensued as indicated by increases in cytokines interleukin 1 beta (IL-1ß) and IL-8 as well as a substantial neutrophil infiltrate. Levels of cJun and JunB proteins, components of the transcription factor AP-1 complex, were also elevated. Up-regulation of extracellular matrix degrading proteinases matrix metalloproteinase 1 (MMP-1), MMP-3, and MMP-9 was noted. A transient increase in keratinocyte proliferation, as indicated by staining for Ki67, was observed. Increased expression of type I and type III procollagen was demonstrated. LIMITATIONS: The data presented are those that resulted from a single treatment session. CONCLUSIONS: Although microablation was confined to the uppermost epidermis, marked changes in epidermal and dermal structure and function were demonstrated after Er:YAG laser microablative resurfacing. We demonstrated substantial dermal matrix remodeling, including a degree of collagen production that compares favorably with some more invasive interventions. Dermal remodeling and stimulation of collagen production are associated with wrinkle reduction. Thus these results suggest that the skin's appearance may be enhanced by creating dermal changes through the use of superficially acting treatments.


Assuntos
Epiderme/patologia , Terapia a Laser/métodos , Lasers de Estado Sólido/uso terapêutico , Metaloproteinase 1 da Matriz/metabolismo , Fator de Transcrição AP-1/metabolismo , Adulto , Idoso , Biomarcadores , Biópsia por Agulha , Proliferação de Células , Citocinas/metabolismo , Procedimentos Cirúrgicos Dermatológicos , Epiderme/metabolismo , Feminino , Seguimentos , Humanos , Imuno-Histoquímica , Queratina-16/metabolismo , Masculino , Metaloproteinase 1 da Matriz/genética , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Complicações Pós-Operatórias/patologia , Ritidoplastia/métodos , Medição de Risco , Estudos de Amostragem , Envelhecimento da Pele , Fatores de Tempo , Resultado do Tratamento , Regulação para Cima
7.
Hum Gene Ther ; 32(9-10): 433-445, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33023309

RESUMO

There is considerable industry excitement about the curative potential of cell and gene therapies, but significant challenges remain in designing cost-effective treatments that are accessible globally. We have taken a modeling-based approach to define the cost and value drivers for cell therapy assets during pharmaceutical drug development. We have created a model development program for a lentiviral modified ex vivo autologous T cell therapy for Oncology indications. Using internal and external benchmarks, we have estimated the total out-of-pocket cost of development for an Oncology cell therapy asset from target identification to filing of marketing application to be $500-600 million. Our model indicates that both clinical and Chemistry Manufacturing and Controls (CMC) cost of development for cell therapies are higher due to unique considerations of ex vivo autologous cell therapies. We have computed a threshold revenue-generating patient number for our model asset that enables selection of assets that can address high unmet medical need and generate pipeline value. Using statistical approaches, we identified that short time to market (<5 years) and reduced commercial cost of goods (<$65,000 per dose) will be essential in developing competitive assets and we propose solutions to reduce both. We emphasize that teams must proactively plan alternate development scenarios with clear articulation of path to value generation and greater patient access. We recommend using a modeling-based approach to enable data driven go/no-go decisions during multigenerational cell therapy development.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Custos de Cuidados de Saúde , Comércio , Terapia Genética , Humanos
8.
Cell Mol Gastroenterol Hepatol ; 8(4): 659-682.e1, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31173894

RESUMO

BACKGROUND & AIMS: Porphyrias are caused by porphyrin accumulation resulting from defects in the heme biosynthetic pathway that typically lead to photosensitivity and possible end-stage liver disease with an increased risk of hepatocellular carcinoma. Our aims were to study the mechanism of porphyrin-induced cell damage and protein aggregation, including liver injury, where light exposure is absent. METHODS: Porphyria was induced in vivo in mice using 3,5-diethoxycarbonyl-1,4-dihydrocollidine or in vitro by exposing human liver Huh7 cells and keratinocytes, or their lysates, to protoporphyrin-IX, other porphyrins, or to δ-aminolevulinic acid plus deferoxamine. The livers, cultured cells, or porphyrin exposed purified proteins were analyzed for protein aggregation and oxidation using immunoblotting, mass spectrometry, and electron paramagnetic resonance spectroscopy. Consequences on cell-cycle progression were assessed. RESULTS: Porphyrin-mediated protein aggregation required porphyrin-photosensitized singlet oxygen and porphyrin carboxylate side-chain deprotonation, and occurred with site-selective native protein methionine oxidation. Noncovalent interaction of protoporphyrin-IX with oxidized proteins led to protein aggregation that was reversed by incubation with acidified n-butanol or high-salt buffer. Phototoxicity and the ensuing proteotoxicity, mimicking porphyria photosensitivity conditions, were validated in cultured keratinocytes. Protoporphyrin-IX inhibited proteasome function by aggregating several proteasomal subunits, and caused cell growth arrest and aggregation of key cell proliferation proteins. Light-independent synergy of protein aggregation was observed when porphyrin was applied together with glucose oxidase as a secondary peroxide source. CONCLUSIONS: Photo-excitable porphyrins with deprotonated carboxylates mediate protein aggregation. Porphyrin-mediated proteotoxicity in the absence of light, as in the liver, requires porphyrin accumulation coupled with a second tissue oxidative injury. These findings provide a potential mechanism for internal organ damage and photosensitivity in porphyrias.


Assuntos
Oxigênio/metabolismo , Porfirias/metabolismo , Ácido Aminolevulínico , Animais , Carcinoma Hepatocelular/metabolismo , Linhagem Celular , Desferroxamina , Heme/metabolismo , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos de Fotossensibilidade , Fármacos Fotossensibilizantes , Porfirias/fisiopatologia , Porfirinas/metabolismo , Agregados Proteicos , Conformação Proteica , Protoporfirinas
9.
J Cell Commun Signal ; 12(1): 401-411, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29086203

RESUMO

In skin, the basement membrane at the dermal-epidermal junction (DEJ-BM) is an important structure that tightly binds the epidermis to the dermis, and acts as a permeability barrier that controls exchange of macromolecules. Repair of the DEJ-BM during wound healing is important for restoration of skin functional properties after wounding. Here, we used a CO2 laser to perform partial thickness wounds in human volunteers, and directly compared wound repair in healthy young and aged individuals, focusing on the DEJ-BM. Our results show that the DEJ-BM is restored within four weeks after partial thickness wounds in young adults. We identified laminin-γ2 as preferred substrate for keratinocytes during reepithelialization of partial thickness human wounds. Laminin-γ2 is expressed continuously by migrating keratinocytes during reepithelialization, whereas collagen IV and collagen VII are deposited after wound closure. In contrast, our study shows that the DEJ-BM restoration following wounding is deficient in elderly individuals. Specifically, COL7A2 was barely increased during wound repair in aged skin and, as a result, the DEJ-BM in elderly skin was not restored and showed abnormal structure. Our data suggest that ameliorating the quality of the DEJ-BM restoration is a promising therapeutic approach to improve the quality of repaired skin in the elderly.

10.
Methods Mol Biol ; 1627: 287-308, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28836209

RESUMO

Type I collagen, or collagen I, is the most abundant protein in the human body and provides strength and resiliency to tissues such as bone, tendons, ligaments, and skin. Collagen I forms macromolecular networks in which resident mesenchymal cells are embedded. Cell-extracellular matrix interactions are critical not only for maintenance of tissue properties but also for guiding and orienting the phenotype of resident cells. Cues from the extracellular matrix have been shown to be critical in pathophysiologies such as fibrosis, aging, and cancer. Hence, the details of these interactions are being scrutinized to better understand the mechanisms of such diseases and conditions. Many in vitro assays, such as cell-embedded collagen lattices, preparation of hydrogels, adhesion assays, etc., have been developed to study various aspects of cell-extracellular matrix interactions. All these in vitro models rely on utilizing high-quality purified collagen I. Here, we provide state-of-the-art collagen I extraction protocol and useful tips to produce high-quality purified collagen I solutions. We also provide a detailed protocol for pepsin digestion of collagen I, for a highly reliable collagen concentration assay, and guidelines for conducting quality controls to validate purified collagen solutions. Collagen I prepared with these procedures is highly suitable for many in vitro applications.


Assuntos
Colágeno Tipo I/isolamento & purificação , Colágeno Tipo I/metabolismo , Cauda , Tendões/metabolismo , Animais , Colágeno Tipo I/química , Concentração de Íons de Hidrogênio , Pepsina A/metabolismo , Proteólise , Ratos , Solubilidade
11.
Methods Mol Biol ; 1627: 395-407, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28836216

RESUMO

The extracellular matrix is critical in guiding cell behavior in normal and pathologic tissues, particularly in cancers and fibrosis. Highlighting the organization (or disorganization) of a collagen-containing matrix can be very useful for understanding or grading pathophysiological conditions. The picrosirius red stain (also called "Sirius red" stain) is one of the best understood histochemical techniques able to selectively highlight collagen networks. Relatively inexpensive, the technique relies on the birefringent properties of collagen molecules. While the picrosirius red stain alone does not selectively bind collagen network, it becomes more specific than the other common collagen stains when combined with polarized light detection. This is why the selective histochemical procedure for collagen detection should be called the picrosirius-polarization method. In this chapter, we will provide essential explanation and detailed protocols and tips to allow collagen researchers not only to better understand how this staining technique works but also to easily apply this technique to their collagen-related research.


Assuntos
Compostos Azo , Colágeno/química , Colágeno/metabolismo , Histocitoquímica , Animais , Birrefringência , Histocitoquímica/métodos , Humanos , Microscopia de Polarização , Ratos , Pele/metabolismo
12.
Methods Mol Biol ; 1627: 117-122, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28836198

RESUMO

Corneal scarring is an obligatory consequence of stroma corneal injury and is a major cause of decreased visual quality and vision loss worldwide. There are currently no satisfactory intervention therapies for corneal fibrosis. In this chapter, we describe well-established in vivo corneal wound models to allow researchers to investigate epithelial and stromal responses to corneal injury.


Assuntos
Cicatriz/etiologia , Cicatriz/patologia , Doenças da Córnea/etiologia , Doenças da Córnea/patologia , Modelos Animais de Doenças , Animais , Cicatriz/diagnóstico , Córnea/patologia , Doenças da Córnea/diagnóstico , Epitélio Corneano/patologia , Camundongos , Microscopia de Fluorescência , Lâmpada de Fenda
13.
Methods Mol Biol ; 1627: 245-251, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28836207

RESUMO

Polyacrylamide hydrogels can be used to culture cells in a range of stiffness that can closer mimic physiological environments. Changes in environmental stiffness have been documented in conditions such as fibrosis, cancer, and aging. In this chapter, we describe a method in which we pour gels directly into multiwell plates using a plastic support that covalently binds to the polymerizing hydrogel. The hydrogel is then crosslinked to calfskin collagen using a crosslinker. The result is a thick hydrogel, scalable to any size plate, which covers the entire surface of the well with no edge effects. The gels can be routinely assembled and are easily reproducible. These scaffolds are used as in vitro models to study fibroblast reaction to variation in environmental stiffness.


Assuntos
Fibroblastos , Hidrogéis , Fenômenos Mecânicos , Resinas Acrílicas , Técnicas de Cultura de Células , Linhagem Celular , Células Cultivadas , Fibroblastos/citologia , Humanos , Hidrogéis/química
14.
Methods Mol Biol ; 1627: 223-233, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28836205

RESUMO

Investigation of cell function is often hampered by the complexity of the tissue context. This problem is circumvented by isolating cells from tissues and analyzing their behavior in culture. Most cell types are cultured as monolayers on planar, rigid Petri dishes, an environment that does not reflect the spatial, three-dimensional cellular environment in vivo. Culture in three-dimensional collagen lattices has been devised to optimize in vitro culture conditions and to provide a more physiologic "in vivo-like" environment. Collagen lattices can easily be manipulated to suit diverse cell types and to provide variable mechanical forces. Cells can be imaged in such surroundings, and gene expression as well as protein production and activity can be monitored.


Assuntos
Técnicas de Cultura de Células , Colágeno/metabolismo , Colágeno/química , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , Biossíntese de Proteínas
15.
Cancer Res ; 77(2): 412-422, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27815387

RESUMO

The lethal phenotype of castration-resistant prostate cancer (CRPC) is generally caused by augmented signaling from the androgen receptor (AR). Here, we report that the AR-repressed gene CCN3/NOV inhibits AR signaling and acts in a negative feedback loop to block AR function. Mechanistically, a cytoplasmic form of CCN3 interacted with the AR N-terminal domain to sequester AR in the cytoplasm of prostate cancer cells, thereby reducing AR transcriptional activity and inhibiting cell growth. However, constitutive repression of CCN3 by the Polycomb group protein EZH2 disrupted this negative feedback loop in both CRPC and enzalutamide-resistant prostate cancer cells. Notably, restoring CCN3 was sufficient to effectively reduce CPRC cell proliferation in vitro and to abolish xenograft tumor growth in vivo Taken together, our findings establish CCN3 as a pivotal regulator of AR signaling and prostate cancer progression and suggest a functional intersection between Polycomb and AR signaling in CRPC. Cancer Res; 77(2); 412-22. ©2016 AACR.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Proteína Sobre-Expressa em Nefroblastoma/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Androgênicos/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Retroalimentação Fisiológica/fisiologia , Xenoenxertos , Humanos , Imunoprecipitação , Masculino , Camundongos , Reação em Cadeia da Polimerase , Neoplasias de Próstata Resistentes à Castração/metabolismo
16.
J Invest Dermatol ; 126(4): 732-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16470170

RESUMO

Retinoids are widely used in the treatment of photoaging to stimulate dermal repair. However, retinoids also induce epidermal hyperplasia, which can lead to excessive scaling. Scaling is the major deterrent to topical retinoid therapy. Keratinocyte growth is strongly stimulated via ligand activation of EGFR. We examined regulation of EGFR ligands by retinoids and the role of EGFR in retinoid-induced hyperplasia in human skin in vivo. Topical treatment of human skin with all-trans retinoic acid (tRA) induces EGFR ligands heparin-binding (HB)-EGF and amphiregulin (AR), and reduces betacellulin mRNA levels. Laser capture microdissection-coupled real-time reverse transcription-PCR reveals that tRA increases HB-EGF mRNA throughout the epidermis, whereas AR induction is limited to basal keratinocytes. Topical tRA activates extracellular signal-regulated kinase 1/2 (Erk1/2) downstream EGFR effectors in human skin in vivo. tRA increases the soluble forms of AR and HB-EGF proteins, and induces epidermal hyperplasia, in human skin organ culture. Neutralization of HB-EGF or AR with specific antibodies strongly reduces tRA-induced epidermal hyperplasia. Finally, inhibition of EGFR activation by genistein reduces epidermal hyperplasia caused by topical retinoid treatment. These data demonstrate the central role of EGFR activation in retinoid-induced epidermal hyperplasia, and suggest that EGFR inhibitors can mitigate retinoid-induced scaling.


Assuntos
Epiderme/efeitos dos fármacos , Epiderme/patologia , Receptores ErbB/agonistas , Retinoides/toxicidade , Dermatopatias/induzido quimicamente , Anfirregulina , Anticorpos/farmacologia , Células Cultivadas , Família de Proteínas EGF , Fator de Crescimento Epidérmico/análise , Fator de Crescimento Epidérmico/antagonistas & inibidores , Fator de Crescimento Epidérmico/metabolismo , Epiderme/química , Glicoproteínas/análise , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Hiperplasia , Peptídeos e Proteínas de Sinalização Intercelular/análise , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ligantes , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Tretinoína/toxicidade
17.
J Cell Commun Signal ; 10(2): 103-20, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27170326

RESUMO

The increased incidence of non-healing skin wounds in developed societies has prompted tremendous research efforts on the complex process known as "wound healing". Unfortunately, the weak relevance of modern wound healing research to human health continues to be a matter of concern. This review summarizes the current knowledge of the cellular mechanisms that mediate wound closure in the skin of humans and laboratory animals. The author highlights the anatomical singularities of human skin vs. the skin of other mammals commonly used for wound healing research (i.e. as mice, rats, rabbits, and pigs), and discusses the roles of stem cells, myofibroblasts, and the matrix environment in the repair process. The majority of this review focuses on reepithelialization and wound closure. Other aspects of wound healing (e.g. inflammation, fibrous healing) are referred to when relevant to the main topic. This review aims at providing the reader with a clear understanding of the similarities and differences that have been reported over the past 100 years between the healing of human wounds and that of other mammals.

18.
Aging Cell ; 15(5): 842-52, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27184009

RESUMO

Human skin heals more slowly in aged vs. young adults, but the mechanism for this delay is unclear. In humans, eccrine sweat glands (ESGs) and hair follicles underlying wounds generate cohesive keratinocyte outgrowths that expand to form the new epidermis. Here, we compared the re-epithelialization of partial-thickness wounds created on the forearm of healthy young (< 40 yo) and aged (> 70 yo) adults. Our results confirm that the outgrowth of cells from ESGs is a major feature of repair in young skin. Strikingly, in aged skin, although ESG density is unaltered, less than 50% of the ESGs generate epithelial outgrowths during repair (vs. 100% in young). Surprisingly, aging does not alter the wound-induced proliferation response in hair follicles or ESGs. Instead, there is an overall reduced cohesiveness of keratinocytes in aged skin. Reduced cell-cell cohesiveness was most obvious in ESG-derived outgrowths that, when present, were surrounded by unconnected cells in the scab overlaying aged wounds. Reduced cell-cell contact persisted during the repair process, with increased intercellular spacing and reduced number of desmosomes. Together, reduced outgrowths of ESG (i) reduce the initial number of cells participating in epidermal repair, (ii) delay wound closure, and (iii) lead to a thinner repaired epidermis in aged vs. young skin. Failure to form cohesive ESG outgrowths may reflect impaired interactions of keratinocytes with the damaged ECM in aged skin. Our findings provide a framework to better understand the mediators of delayed re-epithelialization in aging and further support the importance of ESGs for the repair of human wounds.


Assuntos
Envelhecimento/patologia , Glândulas Écrinas/patologia , Pele/patologia , Cicatrização , Adulto , Idoso , Idoso de 80 Anos ou mais , Proliferação de Células , Desmossomos/metabolismo , Epiderme/patologia , Feminino , Humanos , Masculino
19.
J Invest Dermatol ; 136(2): 444-452, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26802239

RESUMO

The epidermal growth factor receptor (EGFR) and its ligands are essential regulators of epithelial biology, which are often amplified in cancer cells. We have previously shown that shRNA-mediated silencing of one of these ligands, amphiregulin (AREG), results in keratinocyte growth arrest that cannot be rescued by soluble extracellular EGFR ligands. To further explore the functional importance of specific AREG domains, we stably transduced keratinocytes expressing tetracycline-inducible AREG-targeted shRNA with lentiviruses expressing silencing-proof, membrane-tethered AREG cytoplasmic and extracellular domains (AREG-CTD and AREG-ECD), as well as full-length AREG precursor (proAREG). Here we show that growth arrest of AREG-silenced keratinocytes occurs in G2/M and is significantly restored by proAREG and AREG-CTD but not by AREG-ECD. Moreover, the AREG-CTD was sufficient to normalize cell cycle distribution profiles and expression of mitosis-related genes. Our findings uncover an important role of the AREG-CTD in regulating cell division, which may be relevant to tumor resistance to EGFR-directed therapies.


Assuntos
Proliferação de Células/genética , Família de Proteínas EGF/genética , Regulação da Expressão Gênica , Inativação Gênica , Queratinócitos/citologia , Anfirregulina/genética , Animais , Western Blotting , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Receptores ErbB/metabolismo , Citometria de Fluxo , Imunofluorescência , Humanos , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real/métodos , Transdução de Sinais/genética
20.
J Invest Dermatol ; 136(9): 1792-1800, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27312025

RESUMO

In a transcriptome study of lesional psoriatic skin (PP) versus normal skin, we found a coexpressed gene module (N5) enriched 11.5-fold for lipid biosynthetic genes. We also observed fewer visible hairs in PP skin, compared with uninvolved nonlesional psoriatic skin or normal skin (P < 0.0001). To ask whether these findings might be due to abnormalities of the pilosebaceous unit, we carried out three-dimensional morphometric analysis of paired PP and nonlesional psoriatic skin biopsies. Sebaceous glands were markedly atrophic in PP versus nonlesional psoriatic skin (91% average reduction in volume, P = 0.031). Module N5 genes were strongly downregulated in PP versus normal skin (fold change < 0.25, 44.4-fold) and strongly upregulated in sebaceous hyperplasia (fold change > 4, 54.1-fold). The intersection of PP-downregulated and sebaceous hyperplasia-upregulated gene lists generated a gene expression signature consisting solely of module N5 genes, whose expression in PP versus normal skin was inversely correlated with the signature of IL17-stimulated keratinocytes. Despite loss of visible hairs, morphometry identified elongated follicles in PP versus nonlesional psoriatic skin (average 1.7 vs. 1.2 µm, P = 0.020). These results document sebaceous gland atrophy in nonscalp psoriasis, identify a cytokine-regulated set of sebaceous gland signature genes, and suggest that loss of visible hair in PP skin may result from abnormal sebaceous gland function.


Assuntos
Alopecia/patologia , Proteínas de Ciclo Celular/genética , Proteínas Nucleares/genética , Psoríase/patologia , Glândulas Sebáceas/patologia , Adulto , Alopecia/fisiopatologia , Atrofia/diagnóstico , Biópsia por Agulha , Citocinas/metabolismo , Proteínas de Ligação a DNA , Progressão da Doença , Feminino , Regulação da Expressão Gênica/genética , Humanos , Imuno-Histoquímica , Masculino , Psoríase/fisiopatologia , Proteínas de Ligação a RNA , Sensibilidade e Especificidade , Técnicas de Cultura de Tecidos , Transcriptoma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA