Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Res Commun ; 3(8): 1594-1606, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37599786

RESUMEN

Despite recent therapeutic advances, the 5-year survival rate for adults with acute myeloid leukemia (AML) is poor and standard-of-care chemotherapy is associated with significant toxicity, highlighting the need for new therapeutic approaches. Recent work from our group and others established that the G protein-coupled estrogen receptor (GPER) is tumor suppressive in melanoma and other solid tumors. We performed a preliminary screen of human cancer cell lines from multiple malignancies and found that LNS8801, a synthetic pharmacologic agonist of GPER currently in early phase clinical trials, promoted apoptosis in human AML cells. Using human AML cell lines and primary cells, we show that LNS8801 inhibits human AML in preclinical in vitro models, while not affecting normal mononuclear cells. Although GPER is broadly expressed in normal and malignant myeloid cells, this cancer-specific LNS8801-induced inhibition appeared to be independent of GPER signaling. LNS8801 induced AML cell death primarily through a caspase-dependent apoptosis pathway. This was independent of secreted classical death receptor ligands, and instead required induction of reactive oxygen species (ROS) and activation of endoplasmic reticulum (ER) stress response pathways including IRE1α. These studies demonstrate a novel activity of LNS8801 in AML cells and show that targeting ER stress with LNS8801 may be a useful therapeutic approach for AML. Significance: Previous work demonstrated that LNS8801 inhibits cancer via GPER activation, especially in solid tumors. Here we show that LNS8801 inhibits AML via GPER-independent mechanisms that include ROS induction and ER activation.


Asunto(s)
Endorribonucleasas , Leucemia Mieloide Aguda , Adulto , Humanos , Especies Reactivas de Oxígeno , Proteínas Serina-Treonina Quinasas , Leucemia Mieloide Aguda/tratamiento farmacológico , Estrógenos , Estrés del Retículo Endoplásmico
2.
bioRxiv ; 2023 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-36945570

RESUMEN

GPER (G protein-coupled estrogen receptor) has been reported to play roles in several areas of physiology including cancer, metabolic disorders, and cardiovascular disease. However, the understanding of where this receptor is expressed in human tissue is limited due to limited available tools and methodologies that can reliably detect GPER protein. Recently, a highly specific monoclonal antibody against GPER (20H15L21) was developed and is suitable for immunohistochemistry. Using this antibody, we show that GPER protein expression varies markedly between normal human tissue, and also among cancer tissue. As GPER is an emerging therapeutic target for cancer and other diseases, this new understanding of GPER distribution will likely be helpful in design and interpretation of ongoing and future GPER research.

3.
STAR Protoc ; 4(1): 102101, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36853703

RESUMEN

Here, we provide a protocol to model the effects of changes to a small number of cells, such as those arising from a mutation or a virus infection, in stratified epithelia. We describe steps for diluting engineered human keratinocytes into a larger population of unmodified cells and using these cells to grow three-dimensional organotypic cultures. We detail steps to observe effects that are not apparent in homogenous organotypic epithelial cultures by visualizing the localization of modified keratinocytes in epithelial layers. For complete details on the use and execution of this protocol, please refer to Hatterschide et al. (2022).1.


Asunto(s)
Carcinoma de Células Escamosas , Queratinocitos , Humanos , Epitelio , Células Epiteliales , Células Cultivadas
4.
Nat Commun ; 13(1): 7923, 2022 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-36564381

RESUMEN

Human melanocytic nevi (moles) result from a brief period of clonal expansion of melanocytes. As a cellular defensive mechanism against oncogene-induced hyperplasia, nevus-resident melanocytes enter a senescent state of stable cell cycle arrest. Senescent melanocytes can persist for months in mice and years in humans with a risk to escape the senescent state and progress to melanoma. The mechanisms providing prolonged survival of senescent melanocytes remain poorly understood. Here, we show that senescent melanocytes in culture and in nevi express high level of the anti-apoptotic BCL-2 family member BCL-W but remain insensitive to the pan-BCL-2 inhibitor ABT-263. We demonstrate that resistance to ABT-263 is driven by mTOR-mediated enhanced translation of another anti-apoptotic member, MCL-1. Strikingly, the combination of ABT-263 and MCL-1 inhibitors results in synthetic lethality to senescent melanocytes, and its topical application sufficient to eliminate nevi in male mice. These data highlight the important role of redundant anti-apoptotic mechanisms for the survival advantage of senescent melanocytes, and the proof-of-concept for a non-invasive combination therapy for nevi removal.


Asunto(s)
Nevo Pigmentado , Nevo , Neoplasias Cutáneas , Masculino , Humanos , Animales , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Melanocitos/metabolismo , Nevo/metabolismo , Neoplasias Cutáneas/metabolismo
5.
Sci Adv ; 8(35): eabn4007, 2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-36054350

RESUMEN

Melanoma risk is 30 times higher in people with lightly pigmented skin versus darkly pigmented skin. Using primary human melanocytes representing the full human skin pigment continuum and preclinical melanoma models, we show that cell-intrinsic differences between dark and light melanocytes regulate melanocyte proliferative capacity and susceptibility to malignant transformation, independent of melanin and ultraviolet exposure. These differences result from dihydroxyphenylalanine (DOPA), a melanin precursor synthesized at higher levels in melanocytes from darkly pigmented skin. We used both high-throughput pharmacologic and genetic in vivo CRISPR screens to determine that DOPA limits melanocyte and melanoma cell proliferation by inhibiting the muscarinic acetylcholine receptor M1 (CHRM1) signaling. Pharmacologic CHRM1 antagonism in melanoma leads to depletion of c-Myc and FOXM1, both of which are proliferation drivers associated with aggressive melanoma. In preclinical mouse melanoma models, pharmacologic inhibition of CHRM1 or FOXM1 inhibited tumor growth. CHRM1 and FOXM1 may be new therapeutic targets for melanoma.

6.
Cancer Res ; 81(23): 5991-6003, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34706862

RESUMEN

Melanoma and most other cancers occur more frequently and have worse prognosis in males compared with females. Although sex steroids are thought to be involved, classical androgen and estrogen receptors are not detectable in most melanomas. Here we show that testosterone promotes melanoma proliferation by activating ZIP9 (SLC39A9), a zinc transporter that is widely expressed in human melanoma but not intentionally targeted by available therapeutics. This testosterone activity required an influx of zinc, activation of MAPK, and nuclear translocation of YAP. FDA-approved inhibitors of the classical androgen receptor also inhibited ZIP9, thereby antagonizing the protumorigenic effects of testosterone in melanoma. In male mice, androgen receptor inhibitors suppressed growth of ZIP9-expressing melanomas but had no effect on isogenic melanomas lacking ZIP9 or on melanomas in females. These data suggest that ZIP9 might be effectively targeted in melanoma and other cancers by repurposing androgen receptor inhibitors that are currently approved only for prostate cancer. SIGNIFICANCE: Testosterone signaling through ZIP9 mediates some of the sex differences in melanoma, and drugs that target AR can be repurposed to block ZIP9 and inhibit melanoma in males.


Asunto(s)
Antagonistas de Receptores Androgénicos/farmacología , Proteínas de Transporte de Catión/antagonistas & inhibidores , Melanoma/tratamiento farmacológico , Receptores Androgénicos/química , Testosterona/farmacología , Andrógenos/farmacología , Animales , Apoptosis , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Movimiento Celular , Proliferación Celular , Humanos , Masculino , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Factores Sexuales , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Invest Dermatol ; 140(11): 2114-2116, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33099398

RESUMEN

NRAS-driven melanomas frequently develop resistance to MAPK/extracellular signal-regulated kinase kinase inhibitors (MEKis), which limits their therapeutic utility. Nguyen et al. (2020) show that MEKi-resistant cells upregulate phosphoglycerate dehydrogenase (PHGDH), the rate-limiting enzyme in serine synthesis. Suppression of PHGDH rendered cells sensitive to MEKis, suggesting that PHGDH may be a therapeutic target for melanoma.


Asunto(s)
Melanoma , Preparaciones Farmacéuticas , GTP Fosfohidrolasas , Glutatión , Humanos , Melanoma/tratamiento farmacológico , Melanoma/genética , Proteínas de la Membrana , Quinasas de Proteína Quinasa Activadas por Mitógenos , Fosfoglicerato-Deshidrogenasa/genética , Fosfoglicerato-Deshidrogenasa/metabolismo , Serina/metabolismo , Regulación hacia Arriba
8.
Cell Mol Gastroenterol Hepatol ; 10(4): 868-880.e1, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32376419

RESUMEN

BACKGROUND & AIMS: Female sex is associated with lower incidence and improved clinical outcomes for most cancer types including pancreatic ductal adenocarcinoma (PDAC). The mechanistic basis for this sex difference is unknown. We hypothesized that estrogen signaling may be responsible, despite the fact that PDAC lacks classic nuclear estrogen receptors. METHODS: Here we used murine syngeneic tumor models and human xenografts to determine that signaling through the nonclassic estrogen receptor G protein-coupled estrogen receptor (GPER) on tumor cells inhibits PDAC. RESULTS: Activation of GPER with the specific, small molecule, synthetic agonist G-1 inhibited PDAC proliferation, depleted c-Myc and programmed death ligand 1 (PD-L1), and increased tumor cell immunogenicity. Systemically administered G-1 was well-tolerated in PDAC bearing mice, induced tumor regression, significantly prolonged survival, and markedly increased the efficacy of PD-1 targeted immune therapy. We detected GPER protein in a majority of spontaneous human PDAC tumors, independent of tumor stage. CONCLUSIONS: These data, coupled with the wide tissue distribution of GPER and our previous work showing that G-1 inhibits melanoma, suggest that GPER agonists may be useful against a range of cancers that are not classically considered sex hormone responsive and that arise in tissues outside of the reproductive system.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Receptores Acoplados a Proteínas G/agonistas , Animales , Antineoplásicos/uso terapéutico , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico
10.
J Cutan Pathol ; 46(5): 310-316, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30666677

RESUMEN

BACKGROUND: Accurate classification of spitzoid melanocytic lesions is difficult due to overlapping clinical and histopathologic features between Spitz nevi, atypical Spitz tumors (ASTs), and spitzoid melanomas. Expression of p16 (CDKN2A) has been used as a marker of spitzoid lesions. However, its expression may be variable. p15 is a tumor suppressor encoded by CDKN2B, loss of which has been recently shown to promote transition from nevus to melanoma. We sought to determine whether p15 is a useful immunohistochemical marker to distinguish Spitz nevi from spitzoid melanomas and to compare p15 and p16 staining in this population. METHODS: Immunohistochemistry for p15 and p16 was performed on Spitz nevi (n = 19), ASTs (n = 41), and spitzoid melanomas (n = 17). Immunoexpression was categorized by a four-tiered system: 0 (negative), 1+ (weak), 2+ (moderate), 3+ (strong). RESULTS: 3+/strong p15 staining was observed in 68.4% of Spitz nevi, 34.2% of ASTs, and 17.7% of spitzoid melanomas. By contrast, we observed 3+ p16 staining in roughly equivalent percentages of Spitz nevi (57.9%), ASTs (56.1%), and spitzoid melanomas (58.8%). CONCLUSION: These data illustrate that p15 may be more useful than p16 as a biomarker to help distinguish benign from malignant spitzoid lesions.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/biosíntesis , Melanoma , Nevo de Células Epitelioides y Fusiformes , Neoplasias Cutáneas , Femenino , Humanos , Inmunohistoquímica , Masculino , Melanoma/metabolismo , Melanoma/patología , Nevo de Células Epitelioides y Fusiformes/metabolismo , Nevo de Células Epitelioides y Fusiformes/patología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
11.
J Invest Dermatol ; 138(9): 1945-1954, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29526763

RESUMEN

Keratinocytes undergo significant structural remodeling during epidermal differentiation, including a broad transformation of the proteome coupled with a reduction in total cellular biomass. This suggests that intracellular digestion of proteins and organelles is necessary for keratinocyte differentiation. Here, we use both genetic and pharmacologic approaches to demonstrate that autophagy and lysosomal functions are required for keratinocyte differentiation in organotypic human skin. Lysosomal activity was required for mechanistic target of rapamycin signaling and mitochondrial oxidative metabolism. In turn, mitochondrial reactive oxygen species, produced as a natural byproduct of oxidative phosphorylation, were necessary for keratinocyte differentiation. Finally, treatment with exogenous reactive oxygen species rescued the differentiation defect in lysosome-inhibited keratinocytes. These findings highlight a reciprocal relationship between lysosomes and mitochondria, in which lysosomes support mitochondrial metabolism and the associated production of mitochondrial reactive oxygen species. The mitochondrial reactive oxygen species released to the cytoplasm in suprabasal keratinocytes triggers autophagy and lysosome-mediated degradation necessary for epidermal differentiation. As defective lysosome-dependent autophagy is associated with common skin diseases including psoriasis and atopic dermatitis, a better understanding of the role of lysosomes in epidermal homeostasis may guide future therapeutic strategies.


Asunto(s)
Epidermis/metabolismo , Lisosomas/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo , Autofagia , Western Blotting , Diferenciación Celular , Células Cultivadas , Humanos , Fosforilación Oxidativa , Transducción de Señal
12.
Elife ; 72018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29336307

RESUMEN

Female sex and history of prior pregnancies are associated with favorable melanoma outcomes. Here, we show that much of the melanoma protective effect likely results from estrogen signaling through the G protein-coupled estrogen receptor (GPER) on melanocytes. Selective GPER activation in primary melanocytes and melanoma cells induced long-term changes that maintained a more differentiated cell state as defined by increased expression of well-established melanocyte differentiation antigens, increased pigment production, decreased proliferative capacity, and decreased expression of the oncodriver and stem cell marker c-Myc. GPER signaling also rendered melanoma cells more vulnerable to immunotherapy. Systemically delivered GPER agonist was well tolerated, and cooperated with immune checkpoint blockade in melanoma-bearing mice to dramatically extend survival, with up to half of mice clearing their tumor. Complete responses were associated with immune memory that protected against tumor rechallenge. GPER may be a useful, pharmacologically accessible target for melanoma.


Asunto(s)
Antineoplásicos/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/patología , Receptores Acoplados a Proteínas G/agonistas , Transducción de Señal , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Pigmentos Biológicos , Receptores de Estrógenos , Análisis de Supervivencia , Resultado del Tratamiento , Células Tumorales Cultivadas
13.
Am J Pathol ; 186(12): 3094-3099, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27855847

RESUMEN

Most melanomas are driven by BRAF(V600E)-activating mutations, while nevi harboring the same mutations have growth arrest. Although decreased p16 expression has been associated with melanoma formation, in recent work, p15 represented a primary effector of oncogene-induced senescence in nevomelanocytes that was diminished in melanomas. This study determined whether decreased p15 levels represent a general biomarker for the transition from nevus to melanoma. We performed p15 and p16 IHC analyses on a random series of nevi and melanomas. Staining was evaluated and graded for percentage and intensity to determine the H score. For real-time quantitative RT-PCR analysis of p15, RNA was extracted from FFPE sections from 14 nevus and melanoma samples via macrodissection. A two-sided t-test was used to evaluate between-group differences in mean H scores and qΔCt values. p15 Expression was significantly increased in melanocytic nevi compared with melanomas (mean H scores, 254.8 versus 132.3; P < 0.001). On p15 staining, the H score differential was greater than that with p16 staining [122.5 (P < 0.001) and 64.8 (P = 0.055), respectively]. Real-time quantitative RT-PCR analysis revealed a lower mean qΔCt value in melanomas, consistent with lower p15 expression (P = 0.018). Together, these data support the hypothesis that decreased p15 expression is a robust biomarker for distinguishing nevus from melanoma.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Melanoma/metabolismo , Nevo Pigmentado/metabolismo , Nevo/metabolismo , Neoplasias Cutáneas/metabolismo , Biomarcadores de Tumor/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Diagnóstico Diferencial , Humanos , Melanocitos/metabolismo , Melanoma/patología , Mutación , Nevo/patología , Nevo Pigmentado/patología , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/patología
14.
Cell Cycle ; 15(15): 2077-86, 2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27295308

RESUMEN

Proliferation and migration of epidermal keratinocytes are essential for proper cutaneous wound closure after injury. αv integrins and several of their ligands-vitronectin, TGFß and thrombospondin-are up-regulated in healing wounds. However, the role of αv integrins in wound re-epithelialization is unknown. Here, we show that genetic depletion or antibody-mediated blockade of pan-integrin αv, or the specific heterodimer αvß6, in keratinocytes limited epidermal proliferation at the wound edge and prevented re-epithelialization of wounded human organotypic skin both in vivo and in vitro. While we did not observe a migration defect upon αv blockade in vivo, αv was necessary for keratinocyte migration over longer distances in organotypic skin. Integrin αv is required for local activation of latent TGFß, and the wound healing defect in the setting of integrin αv loss was rescued by exogenous, active TGFß, indicating that the αv-TGFß signaling axis is a critical component of the normal epidermal wound healing program. As chronic wounds are associated with decreased TGFß signaling, restoration of TGFß activity may have therapeutic utility in some clinical settings.


Asunto(s)
Epidermis/patología , Integrina alfaV/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas , Animales , Anticuerpos Bloqueadores/farmacología , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Células HEK293 , Humanos , Recién Nacido , Masculino , Ratones SCID , Transducción de Señal/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos
16.
Elife ; 52016 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-27115344

RESUMEN

The association between pregnancy and altered cutaneous pigmentation has been documented for over two millennia, suggesting that sex hormones play a role in regulating epidermal melanocyte (MC) homeostasis. Here we show that physiologic estrogen (17ß-estradiol) and progesterone reciprocally regulate melanin synthesis. This is intriguing given that we also show that normal primary human MCs lack classical estrogen or progesterone receptors (ER or PR). Utilizing both genetic and pharmacologic approaches, we establish that sex steroid effects on human pigment synthesis are mediated by the membrane-bound, steroid hormone receptors G protein-coupled estrogen receptor (GPER), and progestin and adipoQ receptor 7 (PAQR7). Activity of these receptors was activated or inhibited by synthetic estrogen or progesterone analogs that do not bind to ER or PR. As safe and effective treatment options for skin pigmentation disorders are limited, these specific GPER and PAQR7 ligands may represent a novel class of therapeutics.


Asunto(s)
Estrógenos/metabolismo , Melaninas/metabolismo , Progesterona/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Progesterona/metabolismo , Pigmentación de la Piel , Células Cultivadas , Humanos , Melanocitos/metabolismo
17.
Cell Rep ; 14(10): 2313-24, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26947070

RESUMEN

In the absence of low-level ER-to-mitochondrial Ca(2+) transfer, ATP levels fall, and AMPK-dependent, mTOR-independent autophagy is induced as an essential survival mechanism in many cell types. Here, we demonstrate that tumorigenic cancer cell lines, transformed primary human fibroblasts, and tumors in vivo respond similarly but that autophagy is insufficient for survival, and cancer cells die while their normal counterparts are spared. Cancer cell death is due to compromised bioenergetics that can be rescued with metabolic substrates or nucleotides and caused by necrosis associated with mitotic catastrophe during their proliferation. Our findings reveal an unexpected dependency on constitutive Ca(2+) transfer to mitochondria for viability of tumorigenic cells and suggest that mitochondrial Ca(2+) addiction is a feature of cancer cells.


Asunto(s)
Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Acetilcisteína/farmacología , Adenosina Trifosfato/metabolismo , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Humanos , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inhibidores , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Compuestos Macrocíclicos/farmacología , Microscopía por Video , Oxazoles/farmacología , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
18.
Genes Dev ; 30(3): 321-36, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26833731

RESUMEN

Oncogene-induced senescence (OIS) and therapy-induced senescence (TIS), while tumor-suppressive, also promote procarcinogenic effects by activating the DNA damage response (DDR), which in turn induces inflammation. This inflammatory response prominently includes an array of cytokines known as the senescence-associated secretory phenotype (SASP). Previous observations link the transcription-associated methyltransferase and oncoprotein MLL1 to the DDR, leading us to investigate the role of MLL1 in SASP expression. Our findings reveal direct MLL1 epigenetic control over proproliferative cell cycle genes: MLL1 inhibition represses expression of proproliferative cell cycle regulators required for DNA replication and DDR activation, thus disabling SASP expression. Strikingly, however, these effects of MLL1 inhibition on SASP gene expression do not impair OIS and, furthermore, abolish the ability of the SASP to enhance cancer cell proliferation. More broadly, MLL1 inhibition also reduces "SASP-like" inflammatory gene expression from cancer cells in vitro and in vivo independently of senescence. Taken together, these data demonstrate that MLL1 inhibition may be a powerful and effective strategy for inducing cancerous growth arrest through the direct epigenetic regulation of proliferation-promoting genes and the avoidance of deleterious OIS- or TIS-related tumor secretomes, which can promote both drug resistance and tumor progression.


Asunto(s)
Senescencia Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Transducción de Señal/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular , Proliferación Celular , Daño del ADN , Técnicas de Silenciamiento del Gen , Células HEK293 , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Humanos , Inflamación/genética , Células MCF-7 , Proteína de la Leucemia Mieloide-Linfoide/antagonistas & inhibidores , FN-kappa B/metabolismo , Neoplasias/fisiopatología , Fenotipo
19.
J Cell Sci ; 128(21): 3997-4013, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26359297

RESUMEN

Integrins play crucial roles in epithelial adhesion, proliferation, wound healing and cancer. In the epidermis, the roles of many integrin subunits are incompletely defined and mechanistic details regarding their functions are lacking. We performed a multiplexed small hairpin (sh)RNA screen to define roles for each subunit in human organotypic skin. We show that integrin-αv (also known as ITGAV) heterodimers are essential for epidermal generation, with integrin-αv loss driving a keratinocyte G1-S cell cycle block. Surprisingly, integrin αv is not localized within keratinocyte focal adhesions, and instead maintains proliferation by controlling cellular (c)-Myc translation through FAK, p38ß and p90RSK1. These phenotypes depend only on the binding partners of integrin-αv--integrin ß5 and integrin ß6 (also known as ITGB5 and ITGB6, respectively). Through inducible depletion of integrin αv in both normal organotypic epidermis and Ras-driven invasive neoplasia, we show that integrin αv is required for de novo tissue generation and neoplastic invasion but that it is dispensable for epidermal maintenance. Heterodimers of integrin αv with integrin ß5 (integrin αvß5) or integrin ß6 (integrin αvß6) are required to similar extents for neoplastic invasion, thus identifying integrin αvß5 and integrin αvß6 heterodimers as potential therapeutic targets for epidermal squamous cell carcinoma.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Integrina alfaV/metabolismo , Integrinas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Vitronectina/metabolismo , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Fibroblastos/citología , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Adhesiones Focales/fisiología , Humanos , Inmunoprecipitación , Cadenas beta de Integrinas/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Melanocitos/citología , Melanocitos/metabolismo , Piel/citología , Piel/metabolismo
20.
Cancer Discov ; 5(10): 1072-85, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26183406

RESUMEN

UNLABELLED: Deletion of the entire CDKN2B-CDKN2A gene cluster is among the most common genetic events in cancer. The tumor-promoting effects are generally attributed to loss of CDKN2A-encoded p16 and p14ARF tumor suppressors. The degree to which the associated CDKN2B-encoded p15 loss contributes to human tumorigenesis is unclear. Here, we show that CDKN2B is highly upregulated in benign melanocytic nevi, contributes to maintaining nevus melanocytes in a growth-arrested premalignant state, and is commonly lost in melanoma. Using primary melanocytes isolated directly from freshly excised human nevi naturally expressing the common BRAF(V600E)-activating mutation, nevi progressing to melanoma, and normal melanocytes engineered to inducibly express BRAF(V600E), we show that BRAF activation results in reversible, TGFß-dependent, p15 induction that halts proliferation. Furthermore, we engineer human skin grafts containing nevus-derived melanocytes to establish a new, architecturally faithful, in vivo melanoma model, and demonstrate that p15 loss promotes the transition from benign nevus to melanoma. SIGNIFICANCE: Although BRAF(V600E) mutations cause melanocytes to initially proliferate into benign moles, mechanisms responsible for their eventual growth arrest are unknown. Using melanocytes from human moles, we show that BRAF activation leads to a CDKN2B induction that is critical for restraining BRAF oncogenic effects, and when lost, contributes to melanoma.


Asunto(s)
Inhibidor p15 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Melanoma/genética , Melanoma/patología , Nevo/genética , Nevo/patología , Animales , Puntos de Control del Ciclo Celular/genética , Transformación Celular Neoplásica/genética , Cromatina/genética , Cromatina/metabolismo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Inmunohistoquímica , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/metabolismo , Ratones , Mutación , Nevo/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Transducción de Señal , Activación Transcripcional , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...