Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Curr Opin Cardiol ; 39(3): 138-147, 2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38386340

RESUMEN

PURPOSE OF REVIEW: The relationship between metabolism and cardiovascular diseases is complex and bidirectional. Cardiac cells must adapt metabolic pathways to meet biosynthetic demands and energy requirements to maintain contractile function. During cancer, this homeostasis is challenged by the increased metabolic demands of proliferating cancer cells. RECENT FINDINGS: Tumors have a systemic metabolic impact that extends beyond the tumor microenvironment. Lipid metabolism is critical to cancer cell proliferation, metabolic adaptation, and increased cardiovascular risk. Metabolites serve as signals which provide insights for diagnosis and prognosis in cardio-oncology patients. SUMMARY: Metabolic processes demonstrate a complex relationship between cancer cell states and cardiovascular remodeling with potential for therapeutic interventions.


Asunto(s)
Cardiopatías , Neoplasias , Humanos , Neoplasias/complicaciones , Neoplasias/tratamiento farmacológico , Redes y Vías Metabólicas , Metabolismo de los Lípidos , Microambiente Tumoral
2.
Mol Ther ; 31(1): 78-89, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36045587

RESUMEN

Androgen receptor signaling inhibitors (ARSIs) are standard of care for advanced prostate cancer (PCa) patients. Eventual resistance to ARSIs can include the expression of androgen receptor (AR) splice variant, AR-V7, expression as a recognized means of ligand-independent androgen signaling. We demonstrated that interleukin (IL)-6-mediated AR-V7 expression requires bone morphogenic protein (BMP) and CD105 receptor activity in both PCa and associated fibroblasts. Chromatin immunoprecipitation supported CD105-dependent ID1- and E2F-mediated expression of RBM38. Further, RNA immune precipitation demonstrated RBM38 binds the AR-cryptic exon 3 to enable AR-V7 generation. The forced expression of AR-V7 by primary prostatic fibroblasts diminished PCa sensitivity to ARSI. Conversely, downregulation of AR-V7 expression in cancer epithelia and associated fibroblasts was achieved by a CD105-neutralizing antibody, carotuximab. These compelling pre-clinical findings initiated an interventional study in PCa patients developing ARSI resistance. The combination of carotuximab and ARSI (i.e., enzalutamide or abiraterone) provided disease stabilization in four of nine assessable ARSI-refractory patients. Circulating tumor cell evaluation showed AR-V7 downregulation in the responsive subjects on combination treatment and revealed a three-gene panel that was predictive of response. The systemic antagonism of BMP/CD105 signaling can support ARSI re-sensitization in pre-clinical models and subjects that have otherwise developed resistance due to AR-V7 expression.


Asunto(s)
Antagonistas de Receptores Androgénicos , Endoglina , Neoplasias de la Próstata Resistentes a la Castración , Receptores Androgénicos , Humanos , Masculino , Resistencia a Antineoplásicos , Células Neoplásicas Circulantes/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Isoformas de Proteínas , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Proteínas de Unión al ARN , Endoglina/antagonistas & inhibidores , Antagonistas de Receptores Androgénicos/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico
3.
Mol Ther ; 30(2): 672-687, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-34274535

RESUMEN

Triple-negative breast cancer (TNBC) has a high propensity for organ-specific metastasis. However, the underlying mechanisms are not well understood. Here we show that the primary TNBC tumor-derived C-X-C motif chemokines 1/2/8 (CXCL1/2/8) stimulate lung-resident fibroblasts to produce the C-C motif chemokines 2/7 (CCL2/7), which, in turn, activate cholesterol synthesis in lung-colonizing TNBC cells and induce angiogenesis at lung metastatic sites. Inhibiting cholesterol synthesis in lung-colonizing breast tumor cells by pulmonary administration of simvastatin-carrying HER3-targeting nanoparticles reduces angiogenesis and growth of lung metastases in a syngeneic TNBC mouse model. Our findings reveal a novel, chemokine-regulated mechanism for the cholesterol synthesis pathway and a critical role of metastatic site-specific cholesterol synthesis in the pulmonary tropism of TNBC metastasis. The study has implications for the unresolved epidemiological observation that use of cholesterol-lowering drugs has no effect on breast cancer incidence but can unexpectedly reduce breast cancer mortality, suggesting interventions of cholesterol synthesis in lung metastases as an effective treatment to improve survival in individuals with TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Animales , Línea Celular Tumoral , Quimiocinas , Humanos , Pulmón/metabolismo , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética
4.
Subcell Biochem ; 98: 3-14, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35378700

RESUMEN

Eukaryotic cells are capable of internalizing different types of cargo by plasma membrane ruffling and forming vesicles in a process known as endocytosis. The most extensively characterized endocytic pathways are clathrin-coated pits, lipid raft/caveolae-mediated endocytosis, phagocytosis, and macropinocytosis. Macropinocytosis is unique among all the endocytic processes due to its nonselective internalization of extracellular fluid, solutes, and membrane in large endocytic vesicles known as macropinosomes with unique susceptibility toward Na+/H+ exchanger inhibitors. Range of cell types capable of macropinocytosis and known to play important role in different physiological processes, which include antigen presentation, nutrient sensing, migration, and signaling. Understanding the physiological function of macropinocytosis will be helpful in filling the gaps in our knowledge and which can be exploited to develop novel therapeutic targets. In this chapter, we discuss the different molecular mechanisms that initiate the process of macropinocytosis with special emphasis on proteins involved and their diversified role in different cell types.


Asunto(s)
Endocitosis , Pinocitosis , Endocitosis/fisiología , Endosomas , Microdominios de Membrana/metabolismo , Fagocitosis
5.
Proc Natl Acad Sci U S A ; 117(15): 8515-8523, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32238563

RESUMEN

Stromal-epithelial interactions dictate cancer progression and therapeutic response. Prostate cancer (PCa) cells were identified to secrete greater concentration of mitochondrial DNA (mtDNA) compared to noncancer epithelia. Based on the recognized coevolution of cancer-associated fibroblasts (CAF) with tumor progression, we tested the role of cancer-derived mtDNA in a mechanism of paracrine signaling. We found that prostatic CAF expressed DEC205, which was not expressed by normal tissue-associated fibroblasts. DEC205 is a transmembrane protein that bound mtDNA and contributed to pattern recognition by Toll-like receptor 9 (TLR9). Complement C3 was the dominant gene targeted by TLR9-induced NF-κB signaling in CAF. The subsequent maturation complement C3 maturation to anaphylatoxin C3a was dependent on PCa epithelial inhibition of catalase in CAF. In a syngeneic tissue recombination model of PCa and associated fibroblast, the antagonism of the C3a receptor and the fibroblastic knockout of TLR9 similarly resulted in immune suppression with a significant reduction in tumor progression, compared to saline-treated tumors associated with wild-type prostatic fibroblasts. Interestingly, docetaxel, a common therapy for advanced PCa, further promoted mtDNA secretion in cultured epithelia, mice, and PCa patients. The antiapoptotic signaling downstream of anaphylatoxin C3a signaling in tumor cells contributed to docetaxel resistance. The inhibition of C3a receptor sensitized PCa epithelia to docetaxel in a synergistic manner. Tumor models of human PCa epithelia with CAF expanded similarly in mice in the presence or absence of docetaxel. The combination therapy of docetaxel and C3 receptor antagonist disrupted the mtDNA/C3a paracrine loop and restored docetaxel sensitivity.


Asunto(s)
Anafilatoxinas/metabolismo , Fibroblastos Asociados al Cáncer/patología , ADN Mitocondrial/metabolismo , Docetaxel/farmacología , Resistencia a Antineoplásicos , Epitelio/patología , Neoplasias de la Próstata/patología , Animales , Antineoplásicos/farmacología , Apoptosis , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Comunicación Paracrina , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Receptor Toll-Like 9/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Biol Chem ; 295(31): 10560-10561, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32737145

RESUMEN

Cancer cell invasion and metastasis rely on invadopodia, important extensions of the cytoskeleton that initiate degradation of the basement membrane that holds a cell in place. Transforming growth factor-ß (TGF-ß) is well-known to induce breast cancer migration and invasion, but the mechanism by which TGF-ß signaling converts into cell motility is not completely understood. A study from Kiepas et al. revealed a new TGF-ß-dependent role for Src homology/collagen adaptor protein (SHCA) in the initiation of dynamic adhesion complexes involved in the formation of invadopodia. These results highlight new therapeutic opportunities for cancer patients that are not sensitive to HER2 antagonists.


Asunto(s)
Neoplasias de la Mama , Lipoma , Podosomas , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular , Colágeno , Humanos , Podosomas/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Factor de Crecimiento Transformador beta/metabolismo
7.
Oncology (Williston Park) ; 35(3): 119-125, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33818052

RESUMEN

DNA-damage repair (DDR) pathway mutations can sensitize cancer cells to a class of cancer therapeutics known as PARP inhibitors. Given that DDR alterations can be found in up to one-third of advanced prostate cancers, PARP inhibitors have recently been established in treatment-refractory settings. We provide an updated review of the clinical data supporting the 4 PARP inhibitors that have undergone the most investigation thus far in metastatic castrate-resistant prostate cancer (mCRPC). Two of these agents are currently approved for the treatment of DDR-altered mCRPC. We end with a discussion on integration of approved PARP inhibitors into advanced prostate cancer clinical practice.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Humanos , Masculino , Selección de Paciente , Estudios Prospectivos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Ensayos Clínicos Controlados Aleatorios como Asunto
8.
Hepatology ; 70(5): 1582-1599, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31044438

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) enhances the growth and recurrence of colorectal cancer (CRC) liver metastasis. With the rising prevalence of NAFLD, a better understanding of the molecular mechanism underlying NAFLD-associated liver metastasis is crucial. Tumor-associated macrophages (TAMs) constitute a large portion of the tumor microenvironment that promotes tumor growth. NOD-like receptor C4 (NLRC4), a component of an inflammasome complex, plays a role in macrophage activation and interleukin (IL)-1ß processing. We aimed to investigate whether NLRC4-mediated TAM polarization contributes to metastatic liver tumor growth in NAFLD. Wild-type and NLRC4-/- mice were fed low-fat or high-fat diet for 6 weeks followed by splenic injection of mouse CRC MC38 cells. The tumors were analyzed 2 weeks after CRC cell injection. High-fat diet-induced NAFLD significantly increased the number and size of CRC liver metastasis. TAMs and CD206-expressing M2 macrophages accumulated markedly in tumors in the presence of NAFLD. NAFLD up-regulated the expression of IL-1ß, NLRC4, and M2 markers in tumors. In NAFLD, but not normal livers, deletion of NLRC4 decreased liver tumor growth accompanied by decreased M2 TAMs and IL-1ß expression in tumors. Wild-type mice showed increased vascularity and vascular endothelial growth factor (VEGF) expression in tumors with NAFLD, but these were reduced in NLRC4-/- mice. When IL-1 signaling was blocked by recombinant IL-1 receptor antagonist, liver tumor formation and M2-type macrophages were reduced, suggesting that IL-1 signaling contributes to M2 polarization and tumor growth in NAFLD. Finally, we found that TAMs, but not liver macrophages, produced more IL-1ß and VEGF following palmitate challenge. Conclusion: In NAFLD, NLRC4 contributes to M2 polarization, IL-1ß, and VEGF production in TAMs, which promote metastatic liver tumor growth.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas de Unión al Calcio/fisiología , Neoplasias del Colon/patología , Inflamasomas/fisiología , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/secundario , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Animales , Femenino , Interleucina-1beta/fisiología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL
9.
J Biol Chem ; 293(21): 8295-8296, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29802140

RESUMEN

Notch signaling plays critical roles in cancer progression, motivating efforts to identify inhibitors of this pathway. Perron et al. report a small-molecule screen intended to discover compounds that could interfere with the downstream transcription factor HES1. Target validation of their compounds unexpectedly identified a novel HES1-interacting protein, prohibitin 2. This highlights a new mechanism to block Notch signaling and prompting further exploration of HES1 biology.


Asunto(s)
Antineoplásicos/farmacología , Ensayos Analíticos de Alto Rendimiento , Neoplasias/tratamiento farmacológico , Multimerización de Proteína/efectos de los fármacos , Receptores Notch/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Factor de Transcripción HES-1/antagonistas & inhibidores , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Receptores Notch/genética , Receptores Notch/metabolismo , Factor de Transcripción HES-1/genética , Factor de Transcripción HES-1/metabolismo
10.
Gastroenterology ; 155(6): 1985-1998.e5, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30144430

RESUMEN

BACKGROUND & AIMS: Growth, progression, and drug resistance of pancreatic ductal adenocarcinomas (PDACs) have been associated with increased levels and activity of glycogen synthase kinase 3 beta (GSK3B) and histone deacetylases (HDACs). We designed and synthesized molecules that simultaneously inhibit the activities of both enzymes. We tested the effects of one of these molecules, Metavert, in pancreatic cancer cells and mice with pancreatic tumors. METHODS: We tested the ability of Metavert to bind GSK3B and HDACs using surface plasmon resonance. MIA PaCa-2, Bx-PC3, HPAF-II, and HPDE6 cell lines were incubated with different concentrations of Metavert, with or without paclitaxel or gemcitabine, or with other inhibitors of GSK3B and HDACs; cells were analyzed for apoptosis and migration and by immunoblotting, immunofluorescence, and real-time polymerase chain reaction. Krasþ/LSLG12D;Trp53þ/LSLR172H;Pdx-1-Cre (KPC) mice (2 months old) were given injections of Metavert (5 mg/kg, 3 times/week) or vehicle (control). B6.129J mice with tumors grown from UN-KPC961-Luc cells were given injections of Metavert or vehicle. Tumors and metastases were counted and pancreata were analyzed by immunohistochemistry. Glucose metabolism was measured using 13C-glucose tracer and mass spectroscopy and flow cytometry. Cytokine levels in blood samples were measured using multiplexing enzyme-linked immunosorbent assay. RESULTS: Metavert significantly reduced survival of PDAC cells but not nontransformed cells; the agent reduced markers of the epithelial-to-mesenchymal transition and stem cells in PDAC cell lines. Cells incubated with Metavert in combination with irradiation and paclitaxel or gemcitabine had reduced survival compared with cells incubated with either agent alone; Metavert increased killing of drug-resistant PDAC cells by paclitaxel and gemcitabine. PDAC cells incubated with Metavert acquired normalized glucose metabolism. Administration of Metavert (alone or in combination with gemcitibine) to KPC mice or mice with syngeneic tumors significantly increased their survival times, slowed tumor growth, prevented tumor metastasis, decreased tumor infiltration by tumor-associated macrophages, and decreased blood levels of cytokines. CONCLUSIONS: In studies of PDAC cells and 2 mouse models of PDAC, we found a dual inhibitor of GSK3B and HDACs (Metavert) to induce cancer cell apoptosis, reduce migration and expression of stem cell markers, and slow growth of tumors and metastases. Metavert had synergistic effects with gemcitabine.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/secundario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Páncreas/metabolismo , Neoplasias Pancreáticas/genética , Gemcitabina
11.
J Biol Chem ; 290(10): 6574-83, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25596528

RESUMEN

Muscle inflammation is often associated with its expansion. Bladder smooth muscle inflammation-induced cell death is accompanied by hyperplasia and hypertrophy as the primary cause for poor bladder function. In mice, DNA damage initiated by chemotherapeutic drug cyclophosphamide activated caspase 1 through the formation of the NLRP3 complex resulting in detrusor hyperplasia. A cyclophosphamide metabolite, acrolein, caused global DNA methylation and accumulation of DNA damage in a mouse model of bladder inflammation and in cultured bladder muscle cells. In correlation, global DNA methylation and NLRP3 expression was up-regulated in human chronic bladder inflammatory tissues. The epigenetic silencing of DNA damage repair gene, Ogg1, could be reversed by the use of demethylating agents. In mice, demethylating agents reversed cyclophosphamide-induced bladder inflammation and detrusor expansion. The transgenic knock-out of Ogg1 in as few as 10% of the detrusor cells tripled the proliferation of the remaining wild type counterparts in an in vitro co-culture titration experiment. Antagonizing IL-1ß with Anakinra, a rheumatoid arthritis therapeutic, prevented detrusor proliferation in conditioned media experiments as well as in a mouse model of bladder inflammation. Radiation treatment validated the role of DNA damage in the NLRP3-associated caspase 1-mediated IL-1ß secretory phenotype. A protein array analysis identified IGF1 to be downstream of IL-1ß signaling. IL-1ß-induced detrusor proliferation and hypertrophy could be reversed with the use of Anakinra as well as an IGF1 neutralizing antibody. IL-1ß antagonists in current clinical practice can exploit the revealed mechanism for DNA damage-mediated muscular expansion.


Asunto(s)
Hiperplasia/metabolismo , Inflamación/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interleucina-1beta/metabolismo , Músculo Liso/patología , Animales , Apoptosis/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Daño del ADN/efectos de los fármacos , ADN Glicosilasas/metabolismo , Humanos , Hiperplasia/patología , Inflamación/genética , Inflamación/patología , Factor I del Crecimiento Similar a la Insulina/genética , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Ratones , Músculo Liso/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal/efectos de los fármacos , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología
13.
Phytother Res ; 29(6): 805-17, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25787773

RESUMEN

Cancer remains a major health problem worldwide. Among many other factors, two regulatory defects that are present in most cancer cells are constitutive activation of Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway and the induction of indoleamine 2, 3-dioxygenase (IDO), an enzyme that catalyzes tryptophan degradation, through JAK/STAT signaling. Cytokine signaling activates STAT proteins in regulating cell proliferation, differentiation, and survival through modulation of target genes. Many phytochemicals can inhibit both JAK/STAT signaling and IDO expression in antigen-presenting cells by targeting different pathways. Some of the promising phytochemicals that are discussed in this review include resveratrol, cucurbitacin, curcumin, (-)-epigallocatechin gallate, and others. It is now evident that phytochemicals play key roles in inhibition of tumor proliferation and development and provide novel means for therapeutic targeting of cancer.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Quinasas Janus/metabolismo , Neoplasias/metabolismo , Fitoquímicos/farmacología , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Animales , Catequina/análogos & derivados , Catequina/farmacología , Proliferación Celular , Cucurbitacinas/farmacología , Curcumina/farmacología , Modelos Animales de Enfermedad , Humanos , Neoplasias/tratamiento farmacológico , Resveratrol , Estilbenos/farmacología
14.
Cancers (Basel) ; 16(5)2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38473414

RESUMEN

Glutamine, a multifaceted nonessential/conditionally essential amino acid integral to cellular metabolism and immune function, holds pivotal importance in the landscape of cancer therapy. This review delves into the intricate dynamics surrounding both glutamine antagonism strategies and glutamine supplementation within the context of cancer treatment, emphasizing the critical role of glutamine metabolism in cancer progression and therapy. Glutamine antagonism, aiming to disrupt tumor growth by targeting critical metabolic pathways, is challenged by the adaptive nature of cancer cells and the complex metabolic microenvironment, potentially compromising its therapeutic efficacy. In contrast, glutamine supplementation supports immune function, improves gut integrity, alleviates treatment-related toxicities, and improves patient well-being. Moreover, recent studies highlighted its contributions to epigenetic regulation within cancer cells and its potential to bolster anti-cancer immune functions. However, glutamine implementation necessitates careful consideration of potential interactions with ongoing treatment regimens and the delicate equilibrium between supporting normal cellular function and promoting tumorigenesis. By critically assessing the implications of both glutamine antagonism strategies and glutamine supplementation, this review aims to offer comprehensive insights into potential therapeutic strategies targeting glutamine metabolism for effective cancer management.

15.
J Clin Oncol ; 42(2): 228-236, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-37890125

RESUMEN

PURPOSE: Prostate cancer represents the most common cancer diagnosis in Black men and is the second leading cause of cancer death in this population. Multilevel disparities have been well-documented in Black men with prostate cancer and play a role in poorer survival outcomes when compared with White men with prostate cancer. In this review, we highlight the changing trend in disparities for systemic therapy outcomes in Black men diagnosed with metastatic prostate cancer. METHODS: We reviewed data from real-world registries and prospective clinical trials with a particular focus on equal access settings to compare outcomes to systemic therapies between Black and White men with metastatic prostate cancer. RESULTS: In metastatic prostate cancer, there is growing evidence to suggest that Black men may have similar, if not better, outcomes to systemic therapies than White men with advanced disease, as corroborated by prospective studies and clinical trials where health care delivery and follow-up are more likely to be standardized. CONCLUSION: This review illustrates the importance of nonbiological drivers of racial disparities in Black men with advanced prostate cancer. Mitigating barriers to health care access and delivery as well as including participation in clinical trials will be pivotal to ongoing efforts to address disparities in systemic therapy outcomes for Black men with metastatic prostate cancer.


Asunto(s)
Negro o Afroamericano , Disparidades en Atención de Salud , Neoplasias de la Próstata , Humanos , Masculino , Accesibilidad a los Servicios de Salud , Estudios Prospectivos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/diagnóstico , Población Blanca
16.
Cell Death Dis ; 15(2): 152, 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38373993

RESUMEN

Unlike most cancer types, the incidence of esophageal adenocarcinoma (EAC) has rapidly escalated in the western world over recent decades. Using whole genome bisulfite sequencing (WGBS), we identify the transcription factor (TF) FOXM1 as an important epigenetic regulator of EAC. FOXM1 plays a critical role in cellular proliferation and tumor growth in EAC patient-derived organoids and cell line models. We identify ERBB2 as an upstream regulator of the expression and transcriptional activity of FOXM1. Unexpectedly, gene set enrichment analysis (GSEA) unbiased screen reveals a prominent anti-correlation between FOXM1 and immune response pathways. Indeed, syngeneic mouse models show that FOXM1 inhibits the infiltration of CD8+ T cells into the tumor microenvironment. Consistently, FOXM1 suppresses CD8+ T cell chemotaxis in vitro and antigen-dependent CD8+ T cell killing. This study characterizes FOXM1 as a significant EAC-promoting TF and elucidates its novel function in regulating anti-tumor immune response.


Asunto(s)
Adenocarcinoma , Linfocitos T CD8-positivos , Neoplasias Esofágicas , Proteína Forkhead Box M1 , Animales , Humanos , Ratones , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Epigenómica , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/inmunología , Proteína Forkhead Box M1/genética , Proteína Forkhead Box M1/metabolismo , Regulación Neoplásica de la Expresión Génica , Inmunidad , Microambiente Tumoral/inmunología
17.
Am J Pathol ; 181(5): 1573-84, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23022210

RESUMEN

Oncosomes are tumor-derived microvesicles that transmit signaling complexes between cell and tissue compartments. Herein, we show that amoeboid tumor cells export large (1- to 10-µm diameter) vesicles, derived from bulky cellular protrusions, that contain metalloproteinases, RNA, caveolin-1, and the GTPase ADP-ribosylation factor 6, and are biologically active toward tumor cells, endothelial cells, and fibroblasts. We describe methods by which large oncosomes can be selectively sorted by flow cytometry and analyzed independently of vesicles <1 µm. Structures resembling large oncosomes were identified in the circulation of different mouse models of prostate cancer, and their abundance correlated with tumor progression. Similar large vesicles were also identified in human tumor tissues, but they were not detected in the benign compartment. They were more abundant in metastases. Our results suggest that tumor microvesicles substantially larger than exosome-sized particles can be visualized and quantified in tissues and in the circulation, and isolated and characterized using clinically adaptable methods. These findings also suggest a mechanism by which migrating tumor cells condition the tumor microenvironment and distant sites, thereby potentiating advanced disease.


Asunto(s)
Micropartículas Derivadas de Células/patología , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/patología , Factor 6 de Ribosilación del ADP , Animales , Caveolina 1/metabolismo , Línea Celular Tumoral , Micropartículas Derivadas de Células/ultraestructura , Citometría de Flujo , Humanos , Masculino , Ratones , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias de la Próstata/ultraestructura
18.
J Pathol ; 226(1): 17-27, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22069040

RESUMEN

Squamous metaplasia (SQM) is a specific phenotype in response to oestrogen in the prostate and oestrogen receptor (ER) α is required to mediate this response. Previous studies utilizing tissue recombination with seminal vesicle (SV) mesenchyme and prostatic ductal tips from wild type and ERαKO mice suggested that both epithelial and stromal ERα are necessary for SQM. However, tissue recombination is conducted in the renal capsule of immune-deficient mice, in which the microenvironment is different from normal prostate microenvironment in the intact mice. Furthermore, whether the requirement of stromal ERα in the SV for developing SQM is the same as in the prostate is unknown. Therefore, there is a clear need to evaluate the respective roles of ERα in prostate epithelial versus stromal compartments in the intact mouse. Here we generated a mouse model that has selectively lost ERα in either stromal (FSP-ERαKO) or epithelial prostate cells (pes-ERαKO) to determine the requirements of ERα for oestrogen-stimulated prostate proliferation and SQM. Our results indicated that FSP-ERαKO prostates develop full and uniform SQM, which suggests that loss of the majority (~65%) of stromal ERα will not influence oestrogen-mediated SQM. In contrast, loss of epithelial ERα inhibits oestrogen-mediated prostate growth and SQM evidenced by decreasing cytokertin 10 positive squamous cell stratification and differentiation, by reduced ERα protein expression in SQM compared to wild type mice ERα, and by the presence of normal proliferative activities in the oestrogen-treated pes-ERαKO prostates. These in vivo results suggest that epithelial ERα is required for oestrogen-mediated proliferative response and could be an appropriate target for preventing aberrant oestrogen signalling in the prostate.


Asunto(s)
Proliferación Celular , Tejido Conectivo/patología , Epitelio/patología , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Próstata/metabolismo , Próstata/patología , Animales , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Metaplasia , Ratones , Ratones Noqueados
19.
Cancers (Basel) ; 15(11)2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37296976

RESUMEN

Tumorigenesis is a result of cell-intrinsic epigenomic and genomic changes as well as cell-extrinsic factors [...].

20.
Oral Health Prev Dent ; 21(1): 49-60, 2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36794777

RESUMEN

PURPOSE: Periodontal disease is potentially related to certain kinds of cancer. This review aimed to summarize the relationship between periodontal disease and breast cancer, providing some strategies for the clinical treatment and periodontal health care of breast cancer patients. MATERIALS AND METHODS: Systematic reviews, randomised controlled trials, prospective and retrospective clinical studies, case series and reports were collected using search terms entered into the PubMed, Google Scholar and JSTOR databases. RESULTS: Research has provided some evidence that periodontal disease is related to the occurrence and development of breast cancer. Periodontal disease and breast cancer have some common pathogenic factors. Periodontal disease may affect the initiation and development of breast cancer involving microorganisms and inflammation. Periodontal health is affected by radiotherapy, chemotherapy, and endocrine therapy for breast cancer. CONCLUSIONS: Periodontal therapy for breast cancer patients should be performed differently according to the stage of cancer treatment. Adjuvant endocrine treatment (e.g. bisphosphonates) has a great impact on oral treatment. Periodontal therapy contributes to the primary prevention of breast cancer. Periodontal health care of breast cancer patients is worthy of clinician attention.


Asunto(s)
Neoplasias de la Mama , Enfermedades Periodontales , Humanos , Femenino , Neoplasias de la Mama/terapia , Neoplasias de la Mama/tratamiento farmacológico , Estudios Prospectivos , Estudios Retrospectivos , Enfermedades Periodontales/complicaciones , Enfermedades Periodontales/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA