Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Res Sq ; 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38978600

RESUMO

Breastfeeding protects against breast cancer in some women but not others, however the mechanism remains elusive. Lactation requires intense secretory activity of the endoplasmic reticulum (ER) for the production of milk proteins and ER- mitochondria contacts for lipid synthesis. We show that in female mice that share the same nuclear genome (BL/6) but differ in mitochondrial genomes (C57 or NZB), the biological processes engaged during lactation are entirely different at the sub-cellular organization and transcriptional levels resulting in anti-tumorigenic lactation in BL/6C57 females and pro-tumorigenic lactation in BL/6NZB females. Single cell sequencing identified a sub-population of cells, uniquely amplified during lactation in BL/6NZB females, which shares the genetic signature that characterizes post-partum breast cancer (PPBC) in humans relative to matched breast cancers in never pregnant women. Our data indicate that differences in ER and mitochondrial-stress responses during lactation between genotypes inadvertently leads to loss of p53 tumor suppressor function in BL/6NZB females allowing the expansion of the PPBC-like sub-population of cells. Overall, our data reveals the unexpected idiosyncratic nature of lactation and its impacts on the risk of the development of PPBC.

3.
Aging Cell ; 21(10): e13665, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36111352

RESUMO

A major limitation in the use of mouse models in breast cancer research is that most mice develop estrogen receptor-alpha (ERα)-negative mammary tumors, while in humans, the majority of breast cancers are ERα-positive. Therefore, developing mouse models that best mimic the disease in humans is of fundamental need. Here, using an inducible MMTV-rtTA/TetO-NeuNT mouse model, we show that despite being driven by the same oncogene, mammary tumors in young mice are ERα-negative, while they are ERα-positive in aged mice. To further elucidate the mechanisms for this observation, we performed RNAseq analysis and identified genes that are uniquely expressed in aged female-derived mammary tumors. We found these genes to be involved in the activation of the ERα axis of the mitochondrial UPR and the ERα-mediated regulation of XBP-1s, a gene involved in the endoplasmic reticulum UPR. Collectively, our results indicate that aging alters the oncogenic trajectory towards the ERα-positive subtype of breast cancers, and that mammary tumors in aged mice are characterized by the upregulation of multiple UPR stress responses regulated by the ERα.


Assuntos
Receptor alfa de Estrogênio , Receptores de Estrogênio , Idoso , Animais , Carcinogênese/genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Camundongos , Oncogenes , Receptores de Estrogênio/metabolismo , Resposta a Proteínas não Dobradas/genética
4.
EMBO Mol Med ; 14(3): e15504, 2022 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-35107853

RESUMO

Prostate cancer (PCa) clinical management relies heavily on androgen-deprivation therapy (ADT). However, despite experiencing initial clinical benefit, patients getting ADT for non-resectable PCa eventually relapse and develop fatal castration-resistant PCa (CRPC). Multiple mechanisms of acquired resistance to treatment have been reported, including metabolic adaptation (Marine et al, 2020). Notably, activation of the endoplasmic reticulum (ER) unfolded protein response (UPR) has been associated with oncogenic transformation (Hart et al, 2012), tumor progression, metastasis dissemination, and resistance to therapy (Chen & Cubillos-Ruiz, 2021). Targeting different branches of ER UPR has been found to be an effective tool against aggressive PCa (Nguyen et al, 2018; Sheng et al, 2019). Therefore, a better understanding of these pathways may lead to the identification of novel drug targets.


Assuntos
Antagonistas de Androgênios , Neoplasias de Próstata Resistentes à Castração , Antagonistas de Androgênios/farmacologia , Antagonistas de Androgênios/uso terapêutico , Humanos , Masculino , Recidiva Local de Neoplasia , Neoplasias de Próstata Resistentes à Castração/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
Cell Rep ; 38(3): 110254, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35045282

RESUMO

Cancer heterogeneity and evolution are not fully understood. Here, we show that mitochondrial DNA of the normal liver shapes tumor progression, histology, and immune environment prior to the acquisition of oncogenic mutation. Using conplastic mice, we show that mtDNA dictates the expression of the mitochondrial unfolded protein response (UPRmt) in the normal liver. Activation of oncogenic mutations in UPRmt-positive liver increases tumor incidence and histological heterogeneity. Further, in a subset of UPRmt-positive mice, invasive liver cancers develop. RNA sequencing (RNA-seq) analysis of the normal liver reveals that, in this subset, the PAPP-A/DDR2/SNAIL axis of invasion pre-exists along with elevated collagen. Since PAPP-A promotes immune evasion, we analyzed the immune signature and found that their livers are immunosuppressed. Further, the PAPP-A signature identifies the immune exhausted subset of hepatocellular carcinoma (HCC) in humans. Our data suggest that mtDNA of normal liver shapes the entire liver cancer portrait upon acquisition of oncogenic mutations.


Assuntos
Carcinoma Hepatocelular/genética , DNA Mitocondrial/genética , Neoplasias Hepáticas/genética , Resposta a Proteínas não Dobradas/genética , Animais , Carcinoma Hepatocelular/patologia , Feminino , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Plasmática A Associada à Gravidez/metabolismo , Transcriptoma
6.
Aging Cancer ; 2(3): 75-81, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34927079

RESUMO

Aging is a major risk factor of developing breast cancer. Despite the fact that post-menopausal women have lower levels of estrogen, older women have a higher rate of estrogen receptor alpha (ERα) positive breast cancer. Conversely, young women who have elevated levels of estrogen tend to develop ERα negative disease that is associated with higher rate of metastasis. This perspective proposes a unifying model centered around the importance of mitochondrial biology in cancer and aging to explain these observations. Mitochondria are essential for the survival of cancer cells and therefore pathways that maintain the functionality of the mitochondrial network in cancer cells fulfill a critical role in the survival of cancer cells. The ERα and the mitochondrial sirtuin-3 (SIRT3) have been reported to be key players of the mitochondrial unfolded protein response (UPRmt) 1-5. The UPRmt is a complex retrograde signaling cascade that regulates the communication between the mitochondria and the nucleus to restore mitochondrial fitness in response to oxidative stress 5-7. SIRT3 is a major regulator of aging 8. Its level decreases with age and single nucleotide polymorphisms (SNPs) that preserve its expression at higher levels are observed in centenarians 9,10. We propose a model whereby the ERα axis of the UPRmt acts to compensate for the loss of SIRT3 observed with age, and becomes the dominant axis of the UPRmt to maintain the integrity of the mitochondria during transformation, thus explaining the selective advantage of ERα positive luminal cells in breast cancer arising from older women.

7.
Front Cell Dev Biol ; 9: 715923, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34631705

RESUMO

Several studies reported that mitochondrial stress induces cytosolic proteostasis. How mitochondrial stress activates proteostasis in the cytosol remains unclear. However, the cross-talk between the mitochondria and cytosolic proteostasis has far reaching implications for treatment of proteopathies including neurodegenerative diseases. This possibility appears within reach since selected drugs have begun to emerge as being able to stimulate mitochondrial-mediated cytosolic proteostasis. In this review, we focus on studies describing how mitochondrial stress activates proteostasis in the cytosol across multiple model organisms. A model is proposed linking mitochondrial-mediated regulation of cytosolic translation, folding capacity, ubiquitination, and proteasome degradation and autophagy as a multi layered control of cytosolic proteostasis that overlaps with the integrated stress response (ISR) and the mitochondrial unfolded protein response (UPRmt). By analogy to the conductor in an orchestra managing multiple instrumental sections into a dynamically integrated musical piece, the cross-talk between these signaling cascades places the mitochondria as a major conductor of cellular integrity.

8.
Sci Rep ; 11(1): 17003, 2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34417525

RESUMO

Several studies reported that mitochondrial stress induces cytosolic proteostasis in yeast and C. elegans. Notably, inhibition of mitochondrial translation with doxcycyline decreases the toxicity of ß-amyloid aggregates, in a C. elegans. However, how mitochondrial stress activates cytosolic proteostasis remains unclear. Further whether doxycycline has this effect in mammals and in disease relevant tissues also remains unclear. We show here that doxycycline treatment in mice drastically reduces the accumulation of proteins destined for degradation by the proteasome in a CNS region-specific manner. This effect is associated with the activation of the ERα axis of the mitochondrial unfolded protein response (UPRmt), in both males and females. However, sexually dimorphic mechanisms of proteasome activation were observed. Doxycycline also activates the proteasome in fission yeast, where ERα is not expressed. Rather, the ancient ERα-coactivator Mms19 regulates this response in yeast. Our results suggest that the UPRmt initiates a conserved mitochondria-to-cytosol stress signal, resulting in proteasome activation, and that this signal has adapted during evolution, in a sex and tissue specific-manner. Therefore, while our results support the use of doxycycline in the prevention of proteopathic diseases, they also indicate that sex is an important variable to consider in the design of future clinical trials using doxycycline.


Assuntos
Sistema Nervoso Central/metabolismo , Doxiciclina/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Sistema Nervoso Central/efeitos dos fármacos , AMP Cíclico/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Complexo de Endopeptidases do Proteassoma/genética , Saccharomyces cerevisiae/metabolismo , Transcrição Gênica/efeitos dos fármacos , Resposta a Proteínas não Dobradas/efeitos dos fármacos
9.
Endocrinology ; 162(2)2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33269387

RESUMO

Several neurodegenerative disorders are characterized by proteasome dysfunctions leading to protein aggregations and pathogenesis. Since we showed that estrogen receptor alpha (ERα) activates the proteasome, drugs able to stimulate ERα in the central nervous system (CNS) could hold potential for therapeutic intervention. However, the transcriptional effects of selective estrogen receptor modulators (SERMs), such as tamoxifen and raloxifene, can be tissue specific. A direct comparison of the effects of different SERMs on gene transcription in the CNS has never been performed. Here, we report an RNA-seq analysis of the spinal cord treated with estrogen, tamoxifen, or raloxifene. We find stark SERM and sex-specific differences in gene expression profiles in the spinal cord. Notably, raloxifene, but not estrogen or tamoxifen, modulates numerous deubiquitinating enzymes, proteasome subunits and assembly factors, and these effects translate into decreased protein aggregates. In the SOD1-G93A mouse model of amyotrophic lateral sclerosis, we found that even a low dose of raloxifene causes a significant decrease in mutant SOD1 aggregates in the spinal cord, accompanied by a delay in the decline of muscle strength in females, but not in males. These results strongly indicate SERM-selective as well as sex-specific effects, and emphasize the importance of sex as a biological variable to be considered for the careful selection of specific SERM for use in clinical trials for neurodegenerative diseases.


Assuntos
Doenças Neurodegenerativas/tratamento farmacológico , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Cloridrato de Raloxifeno/uso terapêutico , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico , Medula Espinal/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Masculino , Camundongos , Complexo de Endopeptidases do Proteassoma/metabolismo , Cloridrato de Raloxifeno/farmacologia , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Caracteres Sexuais , Medula Espinal/enzimologia , Ubiquitinação/efeitos dos fármacos
10.
J Mammary Gland Biol Neoplasia ; 25(3): 181-189, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32901383

RESUMO

Insulin-like growth factor (IGF) signaling and control of local bioavailability of free IGF by the IGF binding proteins (IGFBP) are important regulators of both mammary development and breast cancer. A recent genome-wide association study (GWAS) identified small nucleotide polymorphisms that reduce the expression of IGFBP-5 as a risk factor of developing breast cancer. This observation suggests that genetic alterations leading to a decreased level of IGFBP-5 may also contribute to breast cancer. In the current review, we focus on Pregnancy-Associated Plasma Protein A (PAPP-A), a protease involved in the degradation of IGFBP-5. PAPP-A is overexpressed in the majority of breast cancers but its role in cancer has only begun to be explored. More specifically, this review aims at highlighting the role of post-partum involution in the oncogenic function of PAPP-A. Notably, we summarize recent studies indicating that PAPP-A plays a role not only in the degradation of IGFBP-5 but also in the deposition of collagen and activation of the collagen receptor discoidin 2 (DDR2) during post-partum involution. Finally, considering the immunosuppressive microenvironment of post-partum involution, we also discuss the unexpected finding made in Ewing Sarcoma that PAPP-A plays a role in immune evasion. While the immunosuppressive role of PAPP-A in breast cancer remains to be determined, collectively these studies highlight the multifaced role of PAPP-A in cancer that extends well beyond its effect on IGF-signaling.


Assuntos
Neoplasias da Mama/genética , Mama/patologia , Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Período Pós-Parto/genética , Proteína Plasmática A Associada à Gravidez/genética , Animais , Mama/fisiopatologia , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Receptor com Domínio Discoidina 2/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Animais/fisiopatologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/fisiopatologia , Camundongos , Proteína Plasmática A Associada à Gravidez/metabolismo , Proteólise , Transdução de Sinais , Somatomedinas/metabolismo , Microambiente Tumoral/genética
11.
EMBO Rep ; 21(4): e48978, 2020 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-32090465

RESUMO

Defects in the proteasome can result in pathological proteinopathies. However, the pathogenic role of sex- and tissue-specific sensitivity to proteotoxic stress remains elusive. Here, we map the proteasome activity across nine tissues, in male and female mice, and demonstrate strong sexual dimorphism in proteasome activity, where females have significantly higher activity in several tissues. Further, we report drastic differences in proteasome activity among tissues, independently of proteasome concentration, which are exacerbated under stress conditions. Sexual dimorphism in proteasome activity is confirmed in a SOD1 ALS mouse model, in which the spinal cord, a tissue with comparatively low proteasome activity, is severely affected. Our results offer mechanistic insight into tissue-specific sensitivities to proteostasis stress and into sex differences in the progression of neurodegenerative proteinopathies.


Assuntos
Esclerose Lateral Amiotrófica , Caracteres Sexuais , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Complexo de Endopeptidases do Proteassoma/genética , Agregados Proteicos , Superóxido Dismutase/genética , Superóxido Dismutase-1/genética
12.
Cancer Res ; 80(9): 1790-1798, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32075799

RESUMO

Childbirth at any age confers a transient increased risk for breast cancer in the first decade postpartum and this window of adverse effect extends over two decades in women with late-age first childbirth (>35 years of age). Crossover to the protective effect of pregnancy is dependent on age at first pregnancy, with young mothers receiving the most benefit. Furthermore, breast cancer diagnosis during the 5- to 10-year postpartum window associates with high risk for subsequent metastatic disease. Notably, lactation has been shown to be protective against breast cancer incidence overall, with varying degrees of protection by race, multiparity, and lifetime duration of lactation. An effect for lactation on breast cancer outcome after diagnosis has not been described. We discuss the most recent data and mechanistic insights underlying these epidemiologic findings. Postpartum involution of the breast has been identified as a key mediator of the increased risk for metastasis in women diagnosed within 5-10 years of a completed pregnancy. During breast involution, immune avoidance, increased lymphatic network, extracellular matrix remodeling, and increased seeding to the liver and lymph node work as interconnected pathways, leading to the adverse effect of a postpartum diagnosis. We al discuss a novel mechanism underlying the protective effect of breastfeeding. Collectively, these mechanistic insights offer potential therapeutic avenues for the prevention and/or improved treatment of postpartum breast cancer.


Assuntos
Neoplasias da Mama/prevenção & controle , Mama/fisiologia , Lactação/fisiologia , Idade Materna , Metástase Neoplásica , Período Pós-Parto/fisiologia , Adulto , Animais , Antígenos CD/metabolismo , Aleitamento Materno , Neoplasias da Mama/química , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/patologia , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Feminino , Proteínas Ligadas por GPI/metabolismo , Saúde Global/estatística & dados numéricos , Humanos , Incidência , Fígado/fisiologia , Camundongos , Proteínas de Neoplasias/metabolismo , Gravidez , Complicações Neoplásicas na Gravidez/diagnóstico , Complicações Neoplásicas na Gravidez/patologia , Proteína Plasmática A Associada à Gravidez/metabolismo , Semaforinas/metabolismo , Fatores de Tempo , Adulto Jovem
13.
Horm Cancer ; 10(4-6): 137-144, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31631239

RESUMO

Pregnancy has a dual effect on the risk of breast cancer. On one hand, pregnancy at a young age is known to be protective. However, pregnancy is also associated with a transient increased risk of breast cancer. For women that have children after the age of 30, the risk remains higher than women who never had children for decades. Involution of the breast has been identified as a window of mammary development associated with the adverse effect of pregnancy. In this review, we summarize the current understanding of the role of involution and describe the role of collagen in this setting. We also discuss the role of a collagen-dependent protease, pappalysin-1, in postpartum breast cancer and its role in activating both insulin-like growth factor signaling and discoidin domain collagen receptor 2, DDR2. Together, these novel advances in our understanding of postpartum breast cancer open the way to targeted therapies against this aggressive breast cancer sub-type.


Assuntos
Neoplasias da Mama/metabolismo , Período Pós-Parto/metabolismo , Proteína Plasmática A Associada à Gravidez/metabolismo , Fatores Etários , Colágeno/metabolismo , Receptor com Domínio Discoidina 2/metabolismo , Feminino , Humanos , Transdução de Sinais , Somatomedinas/metabolismo
14.
Cancer Res ; 79(24): 6057-6066, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31484668

RESUMO

The discovery of the Warburg effect, the preference of cancer cells to generate ATP via glycolysis rather than oxidative phosphorylation, has fostered the misconception that cancer cells become independent of the electron transport chain (ETC) for survival. This is inconsistent with the need of ETC function for the generation of pyrimidines. Along with this misconception, a large body of literature has reported numerous mutations in mitochondrial DNA (mtDNA), further fueling the notion of nonfunctional ETC in cancer cells. More recent findings, however, suggest that cancers maintain oxidative phosphorylation capacity and that the role of mtDNA mutations in cancer is likely far more nuanced in light of the remarkable complexity of mitochondrial genetics. This review aims at describing the various model systems that were developed to dissect the role of mtDNA in cancer, including cybrids, and more recently mitochondrial-nuclear exchange and conplastic mice. Furthermore, we put forward the notion of mtDNA landscapes, where the surrounding nonsynonymous mutations and variants can enhance or repress the biological effect of specific mtDNA mutations. Notably, we review recent studies describing the ability of some mtDNA landscapes to activate the mitochondrial unfolded protein response (UPRmt) but not others. Furthermore, the role of the UPRmt in maintaining cancer cells in the mitohormetic zone to provide selective adaptation to stress is discussed. Among the genes activated by the UPRmt, we suggest that the dismutases SOD2 and SOD1 may play key roles in the establishment of the mitohormetic zone. Finally, we propose that using a UPRmt nuclear gene expression signature may be a more reliable readout than mtDNA landscapes, given their diversity and complexity.


Assuntos
Núcleo Celular/patologia , DNA Mitocondrial/genética , Mitocôndrias/patologia , Neoplasias/genética , Resposta a Proteínas não Dobradas/genética , Animais , Núcleo Celular/genética , DNA Mitocondrial/metabolismo , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Hormese/genética , Humanos , Camundongos , Mitocôndrias/genética , Modelos Biológicos , Mutação , Neoplasias/patologia , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1/metabolismo
15.
Oncogene ; 38(29): 5751-5765, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31222103

RESUMO

We previously reported that the dismutase SOD1 is overexpressed in breast cancer. However, whether SOD1 plays an active role in tumor formation in vivo has never been demonstrated. Further, as luminal cells of normal breast epithelial cells are enriched in SOD1, whether SOD1 is essential for normal mammary gland development has never been determined. We initiated this study to investigate the role of SOD1 in mammary gland tumorigenesis as well as in normal mammary gland development. We crossed the inducible erbB2 (MMTV-iErbB2) and Wnt (MMTV-Wnt) transgenic mice to the SOD1 heterozygote or knockout mice. Our results show that SOD1 is essential for oncogene-driven proliferation, but not normal proliferation of the mammary gland associated with pregnancy or other normal proliferative tissues such as skin and intestines. We show that activation of the oncogene ErbB2 is associated with increased ROS and that high ROS sub-population of ErbB2 cancer cells show elevated SOD1. In the same cells, decrease in SOD1 is associated with an elevation in both apoptosis as well as oncogene-induced senescence. Based on these results, we suggest that SOD1 carries a housekeeping function that maintains ROS levels below a threshold that supports oncogene-dependent proliferation, while allowing escape from oncogene-induced senescence, independently of the oncogene driving tumor formation. These results identify SOD1 as an ideal target for cancer therapy as SOD1 inhibitors hold the potential to prevent the growth of cancers cells of diverse genotypes, activate multiple modes of cell death therefore making acquired resistance more difficult, while sparing normal tissues.


Assuntos
Carcinogênese/genética , Proliferação de Células , Neoplasias Mamárias Animais/genética , Oncogenes , Superóxido Dismutase-1/genética , Animais , Apoptose/genética , Feminino , Genes Essenciais , Humanos , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Transgênicos , Estresse Oxidativo , Gravidez , Receptor ErbB-2/genética , Superóxidos/metabolismo
16.
Breast Cancer Res ; 21(1): 56, 2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-31046834

RESUMO

BACKGROUND: Women who had children at a young age (less than 25) show a reduced overall risk of breast cancer. However, epidemiological studies showed that for all other women, pregnancy increases the risk of breast cancer and the risk remains higher for decades. Further, even in women who had children at a young age, there is a transient increase risk that peaks 6 years after pregnancy. Women diagnosed with breast cancer following pregnancy show a higher rate of metastasis. Yet, the factors that increase the predisposition of post-partum breasts to more aggressive cancers remain unknown. Pregnancy-associated plasma protein A (PAPP-A) is a secreted protease that is overexpressed in more than 70% of breast cancers. However, PAPP-A is a collagen-dependent oncogene. We initiated this study to test the effect of PAPP-A on the predisposition of post-partum breasts. METHODS: We used PAPP-A mouse models for the analysis of its effect on virgin, involuting, or post-partum mammary glands. We performed second-harmonic generation microscopy for the analysis of collagen, defined tumor-associated collagen signature (TACS), the rate of mammary tumors, and the status of the collagen-DDR2-Snail axis of metastasis. We knockdown DDR2 by CRISPR and performed invasion assays. A transcriptomic approach was used to define a PAPP-A and parity-dependent genetic signature and assess its correlation with breast cancer recurrence in humans. RESULTS: We confirmed that post-partum mammary glands have a higher level of collagen than virgin glands and that this collagen is characterized by an anti-proliferative architecture. However, PAPP-A converts the anti-proliferative post-partum collagen into pro-tumorigenic collagen. We show that PAPP-A activates the collagen receptor DDR2 and metastasis. Further, deletion of DDR2 by CRISPR abolished the effect of PAPP-A on invasion. We defined a PAPP-A-driven genetic signature that identifies patients at higher risk of metastasis. CONCLUSIONS: These results support the notion that information about pregnancy may be critical in the prognosis of breast cancer as passage through a single pregnancy predisposes to the oncogenic action of PAPP-A. Our data indicate that history of pregnancy combined with the expression of PAPP-A-driven genetic signature may be useful to identify patients at higher risk of metastatic disease.


Assuntos
Neoplasias da Mama/etiologia , Neoplasias da Mama/metabolismo , Receptor com Domínio Discoidina 2/genética , Regulação da Expressão Gênica , Paridade , Proteína Plasmática A Associada à Gravidez/metabolismo , Animais , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Receptor com Domínio Discoidina 2/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Metástase Neoplásica , Gravidez , Prognóstico , Transdução de Sinais , Fatores de Transcrição da Família Snail/metabolismo
17.
Cell Rep ; 27(8): 2292-2303.e6, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31116976

RESUMO

Moderate mitochondrial stress can lead to persistent activation of cytoprotective mechanisms - a phenomenon termed mitohormesis. Here, we show that mitohormesis primes a subpopulation of cancer cells to basally upregulate mitochondrial stress responses, such as the mitochondrial unfolded protein response (UPRmt) providing an adaptive metastatic advantage. In this subpopulation, UPRmt activation persists in the absence of stress, resulting in reduced oxidative stress indicative of mitohormesis. Mechanistically, we showed that the SIRT3 axis of UPRmt is necessary for invasion and metastasis. In breast cancer patients, a 7-gene UPRmt signature demonstrated that UPRmt-HIGH patients have significantly worse clinical outcomes, including metastasis. Transcriptomic analyses revealed that UPRmt-HIGH patients have expression profiles characterized by metastatic programs and the cytoprotective outcomes of mitohormesis. While mitohormesis is associated with health and longevity in non-pathological settings, these results indicate that it is perniciously used by cancer cells to promote tumor progression.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Proteínas Mitocondriais/metabolismo , Resposta a Proteínas não Dobradas/genética , Animais , Feminino , Humanos , Camundongos , Metástase Neoplásica , Espécies Reativas de Oxigênio
18.
Mol Cell Neurosci ; 98: 12-18, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31028834

RESUMO

The mitochondrial unfolded protein response (UPRmt) is rapidly gaining attention. While the CHOP (ATF4/5) axis of the UPRmt was the first to be described, other axes have subsequently been reported. Validation of this complex pathway in C. elegans has been extensively studied. However, validation of the UPRmt in mouse models of disease known to implicate mitochondrial reprogramming or dysfunction, such as cancer and neurodegeneration, respectively, is only beginning to emerge. This review summarizes recent findings and highlights the major role of the superoxide dismutase SOD1 in the communication between the mitochondria and the nucleus in these settings. While SOD1 has mostly been studied in the context of familial amyotrophic lateral sclerosis (fALS), recent studies suggest that SOD1 may be a potentially important mediator of the UPRmt and converge to emphasize an increasingly vital role of SOD1 as a therapeutic target in cancer.


Assuntos
Mitocôndrias/metabolismo , Neoplasias/metabolismo , Doenças Neurodegenerativas/metabolismo , Superóxido Dismutase-1/metabolismo , Resposta a Proteínas não Dobradas , Animais , Núcleo Celular/metabolismo , Humanos , Estresse Oxidativo , Transdução de Sinais
19.
Nat Commun ; 9(1): 4962, 2018 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-30470740

RESUMO

Activation of the Hippo pathway effector Yap underlies many liver cancers, however no germline or somatic mutations have been identified. Autophagy maintains essential metabolic functions of the liver, and autophagy-deficient murine models develop benign adenomas and hepatomegaly, which have been attributed to activation of the p62/Sqstm1-Nrf2 axis. Here, we show that Yap is an autophagy substrate and mediator of tissue remodeling and hepatocarcinogenesis independent of the p62/Sqstm1-Nrf2 axis. Hepatocyte-specific deletion of Atg7 promotes liver size, fibrosis, progenitor cell expansion, and hepatocarcinogenesis, which is rescued by concurrent deletion of Yap. Our results shed new light on mechanisms of Yap degradation and the sequence of events that follow disruption of autophagy, which is impaired in chronic liver disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagia , Hepatócitos/citologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/fisiopatologia , Fígado/metabolismo , Fosfoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteína 7 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/metabolismo , Carcinogênese , Proteínas de Ciclo Celular , Diferenciação Celular , Feminino , Hepatócitos/metabolismo , Humanos , Fígado/citologia , Fígado/patologia , Neoplasias Hepáticas/genética , Masculino , Camundongos , Fosfoproteínas/genética , Proteólise , Fatores de Transcrição , Proteínas de Sinalização YAP
20.
Front Oncol ; 7: 159, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28798902

RESUMO

Upon accumulation of misfolded proteins in the mitochondria, the mitochondrial unfolded protein response (UPRmt) is activated. This review focuses on the role of this response in cancer. We discuss evidence that during transformation, the UPRmt may play an essential role in the maintenance of the integrity of the mitochondria in the face of increased oxidative stress. However, the role of the UPRmt in other diseases is also emerging and is therefore also briefly discussed.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...