Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Cell ; 159(7): 1615-25, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25525879

RESUMO

Sirtuins (SIRTs) are critical enzymes that govern genome regulation, metabolism, and aging. Despite conserved deacetylase domains, mitochondrial SIRT4 and SIRT5 have little to no deacetylase activity, and a robust catalytic activity for SIRT4 has been elusive. Here, we establish SIRT4 as a cellular lipoamidase that regulates the pyruvate dehydrogenase complex (PDH). Importantly, SIRT4 catalytic efficiency for lipoyl- and biotinyl-lysine modifications is superior to its deacetylation activity. PDH, which converts pyruvate to acetyl-CoA, has been known to be primarily regulated by phosphorylation of its E1 component. We determine that SIRT4 enzymatically hydrolyzes the lipoamide cofactors from the E2 component dihydrolipoyllysine acetyltransferase (DLAT), diminishing PDH activity. We demonstrate SIRT4-mediated regulation of DLAT lipoyl levels and PDH activity in cells and in vivo, in mouse liver. Furthermore, metabolic flux switching via glutamine stimulation induces SIRT4 lipoamidase activity to inhibit PDH, highlighting SIRT4 as a guardian of cellular metabolism.


Assuntos
Proteínas Mitocondriais/metabolismo , Complexo Piruvato Desidrogenase/metabolismo , Sirtuínas/metabolismo , Amidoidrolases/metabolismo , Animais , Técnicas de Silenciamento de Genes , Glutamina/metabolismo , Humanos , Fígado/metabolismo , Camundongos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Ratos , Sirtuínas/genética , Ácido Tióctico/análogos & derivados , Ácido Tióctico/metabolismo
2.
Genes Dev ; 34(21-22): 1422-1438, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-33872192

RESUMO

Breast cancer is the second leading cause of cancer-related death in women and is a complex disease with high intratumoral and intertumoral heterogeneity. Such heterogeneity is a major driving force behind failure of current therapies and development of resistance. Due to the limitations of conventional therapies and inevitable emergence of acquired drug resistance (chemo and endocrine) as well as radio resistance, it is essential to design novel therapeutic strategies to improve the prognosis for breast cancer patients. Deregulated Notch signaling within the breast tumor and its tumor microenvironment (TME) is linked to poor clinical outcomes in treatment of resistant breast cancer. Notch receptors and ligands are also important for normal mammary development, suggesting the potential for conserved signaling pathways between normal mammary gland development and breast cancer. In this review, we focus on mechanisms by which Notch receptors and ligands contribute to normal mammary gland development and breast tumor progression. We also discuss how complex interactions between cancer cells and the TME may reduce treatment efficacy and ultimately lead to acquired drug or radio resistance. Potential combinatorial approaches aimed at disrupting Notch- and TME-mediated resistance that may aid in achieving in an improved patient prognosis are also highlighted.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/fisiopatologia , Neoplasias da Mama/terapia , Resistencia a Medicamentos Antineoplásicos/genética , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Antineoplásicos/farmacologia , Humanos
3.
Genome Res ; 31(2): 337-347, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33361113

RESUMO

Understanding the changes in diverse molecular pathways underlying the development of breast tumors is critical for improving diagnosis, treatment, and drug development. Here, we used RNA-profiling of canine mammary tumors (CMTs) coupled with a robust analysis framework to model molecular changes in human breast cancer. Our study leveraged a key advantage of the canine model, the frequent presence of multiple naturally occurring tumors at diagnosis, thus providing samples spanning normal tissue and benign and malignant tumors from each patient. We showed human breast cancer signals, at both expression and mutation level, are evident in CMTs. Profiling multiple tumors per patient enabled by the CMT model allowed us to resolve statistically robust transcription patterns and biological pathways specific to malignant tumors versus those arising in benign tumors or shared with normal tissues. We showed that multiple histological samples per patient is necessary to effectively capture these progression-related signatures, and that carcinoma-specific signatures are predictive of survival for human breast cancer patients. To catalyze and support similar analyses and use of the CMT model by other biomedical researchers, we provide FREYA, a robust data processing pipeline and statistical analyses framework.

4.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1060-1071, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29309924

RESUMO

Metastatic breast cancer is a leading cause of cancer-related deaths in women worldwide. Patients with triple negative breast cancer (TNBCs), a highly aggressive tumor subtype, have a particularly poor prognosis. Multiple reports demonstrate that altered content of the multicopy mitochondrial genome (mtDNA) in primary breast tumors correlates with poor prognosis. We earlier reported that mtDNA copy number reduction in breast cancer cell lines induces an epithelial-mesenchymal transition associated with metastasis. However, it is unknown whether the breast tumor subtypes (TNBC, Luminal and HER2+) differ in the nature and amount of mitochondrial defects and if mitochondrial defects can be used as a marker to identify tumors at risk for metastasis. By analyzing human primary tumors, cell lines and the TCGA dataset, we demonstrate a high degree of variability in mitochondrial defects among the tumor subtypes and TNBCs, in particular, exhibit higher frequency of mitochondrial defects, including reduced mtDNA content, mtDNA sequence imbalance (mtRNR1:ND4), impaired mitochondrial respiration and metabolic switch to glycolysis which is associated with tumorigenicity. We identified that genes involved in maintenance of mitochondrial structural and functional integrity are differentially expressed in TNBCs compared to non-TNBC tumors. Furthermore, we identified a subset of TNBC tumors that contain lower expression of epithelial splicing regulatory protein (ESRP)-1, typical of metastasizing cells. The overall impact of our findings reported here is that mitochondrial heterogeneity among TNBCs can be used to identify TNBC patients at risk of metastasis and the altered metabolism and metabolic genes can be targeted to improve chemotherapeutic response.


Assuntos
DNA Mitocondrial , Mitocôndrias , Proteínas Mitocondriais , Proteínas de Neoplasias , Proteínas de Ligação a RNA , Neoplasias de Mama Triplo Negativas , Linhagem Celular Tumoral , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Feminino , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
5.
iScience ; 27(4): 109366, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38510127

RESUMO

Triple-negative breast cancer (TNBC) contributes greatly to mortality of breast cancer, demanding new targetable options. We have shown that TNBC patients have high ΔNp63 expression in tumors. However, the function of ΔNp63 in established TNBC is yet to be explored. In current studies, targeting ΔNp63 with inducible CRISPR knockout and Histone deacetylase inhibitor Quisinostat showed that ΔNp63 is important for tumor progression and metastasis in established tumors by promoting myeloid-derived suppressor cell (MDSC) survival through tumor necrosis factor alpha. Decreasing ΔNp63 levels are associated with decreased CD4+ and FOXP3+ T-cells but increased CD8+ T-cells. RNA sequencing analysis indicates that loss of ΔNp63 alters multiple MDSC properties such as lipid metabolism, chemotaxis, migration, and neutrophil degranulation besides survival. We further demonstrated that targeting ΔNp63 sensitizes chemotherapy. Overall, we showed that ΔNp63 reprograms the MDSC-mediated immunosuppressive functions in TNBC, highlighting the benefit of targeting ΔNp63 in chemotherapy-resistant TNBC.

6.
Stem Cells ; 30(7): 1496-508, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22523003

RESUMO

The transcription factor E74-like factor 5 (Elf5) functions downstream of the prolactin receptor signaling pathway and plays an important role in mammary gland development. Using conditional mouse knockouts, we have previously shown that Elf5-null mammary glands exhibit a complete failure of alveologenesis during pregnancy. The Elf5-null developmental phenotype is mediated through alteration in the expression of several critical genes involved in alveologenesis, particularly those belonging to the JAK/STAT pathway. Here, we demonstrate that in addition to regulating terminal differentiation of alveolar cells, Elf5 also plays a critical role in determining cell fate and in regulating the stem/progenitor function of the mammary epithelium. Targeted deletion of Elf5 in the mammary glands leads to accumulation of cell types with dual luminal/basal properties such as coexpression of K8 and K14 and an increase in CD61(+) luminal progenitor population during pregnancy. Further interrogation suggests that the abnormal increase in K14(+) K8(+) cells may represent the CD61(+) luminal progenitors blocked in differentiation. Remarkably, Elf5 deficiency in mammary epithelium also triggers an increase of adult mammary stem activity as evidenced by the accumulation of mammary stem cell (MaSC)-enriched cell population in both pregnant and virgin mice and further confirmed by mammosphere and transplantation assays. Additional support for this phenotype comes from the enriched MaSC gene signature based on transcriptomic analysis of the Elf5-null mammary gland. Finally, our biochemical studies suggest that Elf5 loss leads to hyperactivation of the Notch signaling pathway, which might constitute in part, the underlying molecular mechanism for the altered cell lineage decisions in Elf5-null mammary epithelial cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Glândulas Mamárias Animais/citologia , Receptores Notch/metabolismo , Células-Tronco/citologia , Fatores de Transcrição/metabolismo , Animais , Western Blotting , Células Cultivadas , Proteínas de Ligação a DNA/genética , Feminino , Integrina beta3/metabolismo , Masculino , Glândulas Mamárias Animais/metabolismo , Camundongos , Camundongos Knockout , Gravidez , Reação em Cadeia da Polimerase em Tempo Real , Receptores Notch/genética , Células-Tronco/metabolismo , Fatores de Transcrição/genética
7.
Cancer Res ; 83(13): 2093-2095, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-37403627

RESUMO

Cytokines in the tumor microenvironment can affect tumor growth, progression, and response to therapy, making them compelling therapeutic agents and targets. IFNγ is a pleiotropic cytokine predominantly secreted by immune cells that binds to its receptors IFNGR1 and IFNGR2 on target cells. Multiple clinical trials have investigated the efficacy of IFNγ in combination with other therapies for treating patients with cancer and have shown varying results. Here, we summarize the known effects of IFNγ signaling on tumor cells and explore the possibility of its use in clinical settings.


Assuntos
Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Interferon gama/metabolismo , Proteínas de Transporte , Microambiente Tumoral
8.
Sci Transl Med ; 15(686): eabl4414, 2023 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-36888695

RESUMO

Natural killer (NK) cells are cytotoxic lymphocytes that accumulate within the tumor microenvironment and are generally considered to be antitumorigenic. Using single-cell RNA sequencing and functional analysis of multiple triple-negative breast cancer (TNBC) and basal tumor samples, we observed a unique subcluster of Socs3highCD11b-CD27- immature NK cells that were present only in TNBC samples. These tumor-infiltrating NK cells expressed a reduced cytotoxic granzyme signature and, in mice, were responsible for activating cancer stem cells through Wnt signaling. NK cell-mediated activation of these cancer stem cells subsequently enhanced tumor progression in mice, whereas depletion of NK cells or Wnt ligand secretion from NK cells by LGK-974 decreased tumor progression. In addition, NK cell depletion or inhibition of their function improved anti-programmed cell death ligand 1 (PD-L1) antibody or chemotherapy response in mice with TNBC. Furthermore, tumor samples from patients with TNBC and non-TNBC revealed that increased numbers of CD56bright NK cells were present in TNBC tumors and were correlated to poor overall survival in patients with TNBC. Together, our findings identify a population of protumorigenic NK cells that may be exploited for both diagnostic and therapeutic strategies to improve outcomes for patients with TNBC.


Assuntos
Antineoplásicos , Neoplasias de Mama Triplo Negativas , Humanos , Animais , Camundongos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Antineoplásicos/uso terapêutico , Células Matadoras Naturais , Antígeno B7-H1/metabolismo , Microambiente Tumoral
9.
Methods Mol Biol ; 2429: 485-500, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35507183

RESUMO

Evidence is emerging that cancer cells are arranged as a hierarchy that spans from stem cells to lineage-restricted progenitor cells. The recent development of spheroid cultures with several tissue type has provided new opportunities to assess cancer stem cell (CSC) activity by allowing them to propagate under conditions that resemble the microenvironment for growth of tumors. One tissue type widely used for stem cell investigations is mammary tissue, and the sphere formation assay has been used in both normal mammary tissue and in breast cancer. Here, we describe detailed experimental methodology for generating and propagating spheres from normal mammary tissue and primary breast tumors of mice, patient derived xenografts (PDXs) and breast cancer cell lines. We further describe how these sphere cultures can be employed for coculture assays to assess the effect of tumor microenvironment (TME) on self-renewal ability of CSCs in breast cancer.


Assuntos
Neoplasias da Mama , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Células-Tronco Neoplásicas/metabolismo , Microambiente Tumoral
11.
Nat Commun ; 12(1): 432, 2021 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-33462238

RESUMO

Development of chemoresistance in breast cancer patients greatly increases mortality. Thus, understanding mechanisms underlying breast cancer resistance to chemotherapy is of paramount importance to overcome this clinical challenge. Although activated Notch receptors have been associated with chemoresistance in cancer, the specific Notch ligands and their molecular mechanisms leading to chemoresistance in breast cancer remain elusive. Using conditional knockout and reporter mouse models, we demonstrate that tumor cells expressing the Notch ligand Dll1 is important for tumor growth and metastasis and bear similarities to tumor-initiating cancer cells (TICs) in breast cancer. RNA-seq and ATAC-seq using reporter models and patient data demonstrated that NF-κB activation is downstream of Dll1 and is associated with a chemoresistant phenotype. Finally, pharmacological blocking of Dll1 or NF-κB pathway completely sensitizes Dll1+ tumors to chemotherapy, highlighting therapeutic avenues for chemotherapy resistant breast cancer patients in the near future.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Proteínas de Ligação ao Cálcio/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas de Membrana/metabolismo , Subunidade p50 de NF-kappa B/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzamidas/farmacologia , Benzamidas/uso terapêutico , Mama/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Conjuntos de Dados como Assunto , Modelos Animais de Doenças , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Subunidade p50 de NF-kappa B/antagonistas & inibidores , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , RNA-Seq , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
12.
Cancer Res ; 81(9): 2399-2414, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33514514

RESUMO

Inflammatory breast cancer (IBC) is a highly metastatic breast carcinoma with high frequency of estrogen receptor α (ERα) negativity. Here we explored the role of the second ER subtype, ERß, and report expression in IBC tumors and its correlation with reduced metastasis. Ablation of ERß in IBC cells promoted cell migration and activated gene networks that control actin reorganization, including G-protein-coupled receptors and downstream effectors that activate Rho GTPases. Analysis of preclinical mouse models of IBC revealed decreased metastasis of IBC tumors when ERß was expressed or activated by chemical agonists. Our findings support a tumor-suppressive role of ERß by demonstrating the ability of the receptor to inhibit dissemination of IBC cells and prevent metastasis. On the basis of these findings, we propose ERß as a potentially novel biomarker and therapeutic target that can inhibit IBC metastasis and reduce its associated mortality. SIGNIFICANCE: These findings demonstrate the capacity of ERß to elicit antimetastatic effects in highly aggressive inflammatory breast cancer and propose ERß and the identified associated genes as potential therapeutic targets in this disease.


Assuntos
Actinas/metabolismo , Movimento Celular/genética , Receptor beta de Estrogênio/metabolismo , Neoplasias Inflamatórias Mamárias/metabolismo , Transdução de Sinais/genética , Citoesqueleto de Actina/metabolismo , Animais , Estudos de Coortes , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Feminino , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Neoplasias Inflamatórias Mamárias/genética , Neoplasias Inflamatórias Mamárias/patologia , Células MCF-7 , Camundongos , Metástase Neoplásica/genética , Transfecção , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Dev Biol ; 329(2): 227-41, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19269284

RESUMO

The transcription factor Elf5 plays an important role in mammary gland development. However, because of the embryonic lethality of Elf5 straight knockout mice, prior studies have been limited to experiments with Elf5 haploinsufficient animals, overexpression systems or transplants. Here, we have utilized K14-Cre to generate mammary-gland specific Elf5 conditional knockout mice. During pregnancy, Elf5-null mammary epithelium completely failed to initiate alveologenesis, and a characteristic of virgin ductal epithelial cells persisted postpartum. We demonstrate that the loss of Elf5 leads to the absence of alveolar secretory markers confirming previous published data. Interestingly, the developmental block due to a lack of Elf5 could not be restored by multiple gestations. Elf5-null mammary epithelial cells also display disorganized cell structures as evident by altered cell polarities, which might be the cause for collapsed lumina. We observe reduced levels of Stat5 and attenuated Stat5 activity as measured by p-Stat5 levels both in Elf5-null mammary glands as well as cultured mammary epithelial cells. This data suggests that the failure of alveolar and lactogenic differentiation due to the loss of Elf5 is mediated in part due to impaired Stat5 activity. In support of this hypothesis, we show by ChIP experiments that Stat5a promoter contains a conserved Elf5-binding site that is occupied by Elf5 in mammary glands. Mammary epithelia lacking Elf5 exhibited downregulation of several other critical genes involved in alveologenesis, suggesting Elf5 as a master regulator in alveolar development. We propose a model for Elf5-mediated alveolar development, in which Elf5 regulates the expression of key mediators of the PrlR/Jak2/Stat5 signaling pathway.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Glândulas Mamárias Animais/embriologia , Fator de Transcrição STAT5/fisiologia , Fatores de Transcrição/fisiologia , Animais , Sequência de Bases , Primers do DNA , Proteínas de Ligação a DNA/genética , Regulação para Baixo/fisiologia , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Fatores de Transcrição/genética , Transcrição Gênica/fisiologia
14.
BMC Mol Biol ; 11: 68, 2010 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-20831799

RESUMO

BACKGROUND: The ETS transcription factor Elf5 (also known as ESE-2) is highly expressed in the mammary gland and plays an important role in its development and differentiation. Indeed studies in mice have illustrated an essential role for Elf5 in directing alveologenesis during pregnancy. Although the molecular mechanisms that underlie the developmental block in Elf5 null mammary glands are beginning to be unraveled, this investigation has been hampered by limited information about the identity of Elf5-target genes. To address this shortcoming, in this study we have performed ChIP-cloning experiments to identify the specific genomic segments that are occupied by Elf5 in pregnant mouse mammary glands. RESULTS: Sequencing and genomic localization of cis-regulatory regions bound by Elf5 in vivo has identified several potential target genes covering broad functional categories. A subset of these target genes demonstrates higher expression levels in Elf5-null mammary glands suggesting a repressive functional role for this transcription factor. Here we focus on one putative target of Elf5, the Ccnd2 gene that appeared in our screen. We identify a novel Elf5-binding segment upstream of the Ccnd2 gene and demonstrate that Elf5 can transcriptionally repress Ccnd2 by directly binding to the proximal promoter region. Finally, using Elf5-null mammary epithelial cells and mammary glands, we show that loss of Elf5 in vivo leads to up regulation of Ccnd2 and an altered expression pattern in luminal cells. CONCLUSIONS: Identification of Elf5-targets is an essential first step in elucidating the transcriptional landscape that is shaped by this important regulator. Our studies offer new toolbox in examining the biological role of Elf5 in mammary gland development and differentiation.


Assuntos
Ciclina D2/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Glândulas Mamárias Animais/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Imunoprecipitação da Cromatina , Ciclina D2/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Sequências Reguladoras de Ácido Nucleico
15.
FEBS Lett ; 594(6): 973-985, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31794060

RESUMO

The ∆Np63 isoform of the p53-family transcription factor Trp63 is a key regulator of mammary epithelial stem cells that is involved in breast cancer development. To investigate the role of ∆Np63 at different stages of normal mammary gland development, we generated a ∆Np63-inducible conditional knockout (cKO) mouse model. We demonstrate that the deletion of ∆Np63 at puberty results in depletion of mammary stem cell-enriched basal cells, reduces expression of E-cadherin and ß-catenin, and leads to a closed ductal lumen. RNA-sequencing analysis reveals reduced expression of oxidative phosphorylation (OXPHOS)-associated proteins and desmosomal polarity proteins. Functional assays show reduced numbers of mitochondria in the mammary epithelial cells of ΔNp63 cKO compared to wild-type, supporting the reduced OXPHOS phenotype. These findings identify a novel role for ∆Np63 in cellular metabolism and mammary epithelial cell polarity.


Assuntos
Polaridade Celular , Glândulas Mamárias Animais/metabolismo , Maturidade Sexual , Células-Tronco/metabolismo , Transativadores/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Feminino , Camundongos , Camundongos Knockout , Transativadores/genética , beta Catenina/genética , beta Catenina/metabolismo
16.
Nat Cell Biol ; 22(5): 591-602, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32284542

RESUMO

Triple-negative breast cancer (TNBC) is characterized by a high degree of immune infiltrate in the tumour microenvironment, which may influence the fate of TNBC cells. We reveal that loss of the tumour suppressive transcription factor Elf5 in TNBC cells activates intrinsic interferon-γ (IFN-γ) signalling, promoting tumour progression and metastasis. Mechanistically, we find that loss of the Elf5-regulated ubiquitin ligase FBXW7 ensures stabilization of its putative protein substrate IFN-γ receptor 1 (IFNGR1) at the protein level in TNBC. Elf5low tumours show enhanced IFN-γ signalling accompanied by an increase of immunosuppressive neutrophils within the tumour microenvironment and increased programmed death ligand 1 expression. Inactivation of either programmed death ligand 1 or IFNGR1 elicited a robust anti-tumour and/or anti-metastatic effect. A positive correlation between ELF5 and FBXW7 expression and a negative correlation between ELF5, FBXW7 and IFNGR1 expression in the tumours of patients with TNBC strongly suggest that this signalling axis could be exploited for patient stratification and immunotherapeutic treatment strategies for Elf5low patients with TNBC.


Assuntos
Proliferação de Células/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteína 7 com Repetições F-Box-WD/metabolismo , Interferon gama/metabolismo , Metástase Neoplásica/patologia , Receptores de Interferon/metabolismo , Fatores de Transcrição/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais/fisiologia , Microambiente Tumoral/fisiologia , Receptor de Interferon gama
17.
Dev Growth Differ ; 51(5): 511-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19469787

RESUMO

Our previous study documented a reproductive function for the male-transmitted mitochondrial DNA (mtDNA)-encoded cytochrome c oxidase subunit II (MCOX2) protein in a unionoid bivalve. Here, immunoblotting, immunohistochemistry and immunoelectron microscopy analyses demonstrate that the female-transmitted protein (FCOX2) is: (i) expressed in both male and female gonads; (ii) maximally expressed in ovaries just prior to the time of the annual fertilization event; (iii) displayed in the cytoplasm and more strongly in the plasma membrane (microvilli), vitelline matrix and vitelline envelope of mature ovarian eggs; and (iv) strongly localized to the vitelline matrix of some eggs just prior to fertilization. These findings represent evidence for the extra-mitochondrial localization of an mtDNA-encoded gene product and are consistent with multifunctionality for FCOX2 in eggs.


Assuntos
DNA Mitocondrial/fisiologia , Complexo IV da Cadeia de Transporte de Elétrons/fisiologia , Reprodução/fisiologia , Unionidae/enzimologia , Unionidae/metabolismo , Sequência de Aminoácidos , Animais , Membrana Celular/metabolismo , DNA Mitocondrial/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Gônadas/metabolismo , Gônadas/ultraestrutura , Immunoblotting , Imuno-Histoquímica , Masculino , Microscopia Imunoeletrônica , Dados de Sequência Molecular , Óvulo/metabolismo , Óvulo/ultraestrutura , Reprodução/genética , Estações do Ano , Unionidae/ultraestrutura
18.
J Clin Med ; 8(5)2019 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-31075939

RESUMO

Epithelial-to-mesenchymal transition (EMT) is a process through which epithelial cells lose their epithelial characteristics and cell-cell contact, thus increasing their invasive potential. In addition to its well-known roles in embryonic development, wound healing, and regeneration, EMT plays an important role in tumor progression and metastatic invasion. In breast cancer, EMT both increases the migratory capacity and invasive potential of tumor cells, and initiates protumorigenic alterations in the tumor microenvironment (TME). In particular, recent evidence has linked increased expression of EMT markers such as TWIST1 and MMPs in breast tumors with increased immune infiltration in the TME. These immune cells then provide cues that promote immune evasion by tumor cells, which is associated with enhanced tumor progression and metastasis. In the current review, we will summarize the current knowledge of the role of EMT in the biology of different subtypes of breast cancer. We will further explore the correlation between genetic switches leading to EMT and EMT-induced alterations within the TME that drive tumor growth and metastasis, as well as their possible effect on therapeutic response in breast cancer.

19.
FEBS Lett ; 593(10): 1030-1039, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31002388

RESUMO

Elf5 is a transcription factor known to regulate critical developmental processes and has been shown to act as a tumour suppressor in multiple cancers. Elf5 knockout mice are embryonically lethal, limiting in vivo studies pertaining to its function. Moreover, haploinsufficiency of Elf5 limits the use of current mouse models to investigate adult tissue distribution of Elf5. Here, we successfully generated Elf5CreERT2-GFP bacterial artificial chromosome (BAC) transgenic mice and show that Elf5+ cells are present in several adult tissues, where its expression was previously not known. Our study demonstrates the unique distribution of Elf5+ cells in multiple adult organs, which will facilitate future studies investigating the function of Elf5 in these tissues during homeostasis, repair and cancer.


Assuntos
Linhagem da Célula , Proteínas de Ligação a DNA/genética , Camundongos Transgênicos , Modelos Animais , Fatores de Transcrição/genética , Animais , Cromossomos Artificiais Bacterianos , Integrases , Camundongos , Camundongos Knockout
20.
Oncogene ; 38(12): 2092-2107, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30442981

RESUMO

Aberrant Notch signaling is implicated in several cancers, including breast cancer. However, the mechanistic details of the specific receptors and function of ligand-mediated Notch signaling that promote breast cancer remains elusive. In our studies we show that DLL1, a Notch signaling ligand, is significantly overexpressed in ERα+ luminal breast cancer. Intriguingly, DLL1 overexpression correlates with poor prognosis in ERα+ luminal breast cancer, but not in other subtypes of breast cancer. In addition, this effect is specific to DLL1, as other Notch ligands (DLL3, JAGGED1, and JAGGED2) do not influence the clinical outcome of ERα+ patients. Genetic studies show that DLL1-mediated Notch signaling in breast cancer is important for tumor cell proliferation, angiogenesis, and cancer stem cell function. Consistent with prognostic clinical data, we found the tumor-promoting function of DLL1 is exclusive to ERα+ luminal breast cancer, as loss of DLL1 inhibits both tumor growth and lung metastasis of luminal breast cancer. Importantly, we find that estrogen signaling stabilizes DLL1 protein by preventing its proteasomal and lysososmal degradations. Moreover, estrogen inhibits ubiquitination of DLL1. Together, our results highlight an unexpected and novel subtype-specific function of DLL1 in promoting luminal breast cancer that is regulated by estrogen signaling. Our studies also emphasize the critical role of assessing subtype-specific mechanisms driving tumor growth and metastasis to generate effective subtype-specific therapeutics.


Assuntos
Neoplasias da Mama/patologia , Estrogênios/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/metabolismo , Proteínas de Ligação ao Cálcio , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica , Progressão da Doença , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Lisossomos/metabolismo , Camundongos , Metástase Neoplásica , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica , Prognóstico , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa