Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
2.
Cells ; 12(23)2023 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-38067144

RESUMEN

The transforming growth factor ß (TGFß) superfamily, consisting of protein ligands, receptors, and intracellular SMAD transducers, regulates fundamental biological processes and cancer development. Our previous study has shown that sustained activation of TGFß receptor 1 (TGFBR1) driven by anti-Mullerian hormone receptor type 2 (Amhr2)-Cre in the mouse testis induces the formation of testicular granulosa cell tumors (TGCTs). As Amhr2-Cre is expressed in both Sertoli cells and Leydig cells, it remains unclear whether the activation of TGFBR1 in Sertoli cells alone is sufficient to induce TGCT formation. Therefore, the objective of this study was to determine whether Sertoli cell-activation of TGFBR1 drives oncogenesis in the testis. Our hypothesis was that overactivation of TGFBR1 in Sertoli cells would promote their transdifferentiation into granulosa-like cells and the formation of TGCTs. To test this hypothesis, we generated mice harboring constitutive activation of TGFBR1 in Sertoli cells using anti-Mullerian hormone (Amh)-Cre. Disorganized seminiferous tubules and tumor nodules were found in TGFBR1CA; Amh-Cre mice. A histological analysis showed that Sertoli cell-specific activation of TGFBR1 led to the development of neoplasms resembling granulosa cell tumors, which derailed spermatogenesis. Moreover, TGCTs expressed granulosa cell markers including FOXL2, FOXO1, and INHA. Using a dual fluorescence reporter line, the membrane-targeted tdTomato (mT)/membrane-targeted EGFP (mG) mouse, we provided evidence that Sertoli cells transdifferentiated toward a granulosa cell fate during tumorigenesis. Thus, our findings indicate that Sertoli cell-specific activation of TGFBR1 leads to the formation of TGCTs, supporting a key contribution of Sertoli cell reprogramming to the development of this testicular malignancy in our model.


Asunto(s)
Tumor de Células de la Granulosa , Neoplasias Ováricas , Neoplasias Testiculares , Masculino , Humanos , Femenino , Ratones , Animales , Células de Sertoli/metabolismo , Tumor de Células de la Granulosa/metabolismo , Tumor de Células de la Granulosa/patología , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Neoplasias Testiculares/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Hormona Antimülleriana/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Neoplasias Ováricas/patología
4.
Am J Pathol ; 193(3): 259-274, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36521562

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic human disease with persistent destruction of lung parenchyma. Transforming growth factor-ß1 (TGF-ß1) signaling plays a pivotal role in the initiation and pathogenesis of IPF. As shown herein, TGF-ß1 signaling down-regulated not only peroxisome biogenesis but also the metabolism of these organelles in human IPF fibroblasts. In vitro cell culture observations in human fibroblasts and human lung tissue indicated that peroxisomal biogenesis and metabolic proteins were significantly down-regulated in the lung of 1-month-old transgenic mice expressing a constitutively active TGF-ß type I receptor kinase (ALK5). The peroxisome biogenesis protein peroxisomal membrane protein Pex13p (PEX13p) as well as the peroxisomal lipid metabolic enzyme peroxisomal acyl-coenzyme A oxidase 1 (ACOX1) and antioxidative enzyme catalase were highly up-regulated in TGF-ß type II receptor and Smad3 knockout mice. This study reports a novel mechanism of peroxisome biogenesis and metabolic regulation via TGF-ß1-Smad signaling: interaction of the Smad3 transcription factor with the PEX13 gene in chromatin immunoprecipitation-on-chip assay as well as in a bleomycin-induced pulmonary fibrosis model applied to TGF-ß type II receptor knockout mice. Taken together, data from this study suggest that TGF-ß1 participates in regulation of peroxisomal biogenesis and metabolism via Smad-dependent signaling, opening up novel strategies for the development of therapeutic approaches to inhibit progression of pulmonary fibrosis patients with IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática , Factor de Crecimiento Transformador beta1 , Ratones , Animales , Humanos , Lactante , Factor de Crecimiento Transformador beta1/metabolismo , Ratones Transgénicos , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/patología , Bleomicina/efectos adversos , Fibroblastos/metabolismo , Ratones Noqueados
5.
Nat Commun ; 13(1): 7551, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36477656

RESUMEN

The pro-tumourigenic role of epithelial TGFß signalling in colorectal cancer (CRC) is controversial. Here, we identify a cohort of born to be bad early-stage (T1) colorectal tumours, with aggressive features and a propensity to disseminate early, that are characterised by high epithelial cell-intrinsic TGFß signalling. In the presence of concurrent Apc and Kras mutations, activation of epithelial TGFß signalling rampantly accelerates tumourigenesis and share transcriptional signatures with those of the born to be bad T1 human tumours and predicts recurrence in stage II CRC. Mechanistically, epithelial TGFß signalling induces a growth-promoting EGFR-signalling module that synergises with mutant APC and KRAS to drive MAPK signalling that re-sensitise tumour cells to MEK and/or EGFR inhibitors. Together, we identify epithelial TGFß signalling both as a determinant of early dissemination and a potential therapeutic vulnerability of CRC's with born to be bad traits.


Asunto(s)
Apoptosis , Factor de Crecimiento Transformador beta , Humanos , Apoptosis/genética
6.
Commun Biol ; 5(1): 1068, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36207615

RESUMEN

TGF-ß signaling is involved in pancreatic ductal adenocarcinoma (PDAC) tumorigenesis, representing one of the four major pathways genetically altered in 100% of PDAC cases. TGF-ß exerts complex and pleiotropic effects in cancers, notably via the activation of SMAD pathways, predominantly SMAD2/3/4. Though SMAD2 and 3 are rarely mutated in cancers, SMAD4 is lost in about 50% of PDAC, and the role of SMAD2/3 in a SMAD4-null context remains understudied. We herein provide evidence of a SMAD2/3 oncogenic effect in response to TGF-ß1 in SMAD4-null human PDAC cancer cells. We report that inactivation of SMAD2/3 in SMAD4-negative PDAC cells compromises TGF-ß-driven collective migration mediated by FAK and Rho/Rac signaling. Moreover, RNA-sequencing analyses highlight a TGF-ß gene signature related to aggressiveness mediated by SMAD2/3 in the absence of SMAD4. Using a PDAC patient cohort, we reveal that SMAD4-negative tumors with high levels of phospho-SMAD2 are more aggressive and have a poorer prognosis. Thus, loss of SMAD4 tumor suppressive activity in PDAC leads to an oncogenic gain-of-function of SMAD2/3, and to the onset of associated deleterious effects.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Proteína smad3/metabolismo , Carcinogénesis/genética , Carcinoma Ductal Pancreático/metabolismo , Humanos , Neoplasias Pancreáticas/metabolismo , ARN , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Neoplasias Pancreáticas
7.
Cancers (Basel) ; 14(9)2022 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-35565312

RESUMEN

Ovarian granulosa cell tumors (GCTs) are rare sex cord-stromal tumors, accounting for ~5% ovarian tumors. The etiology of GCTs remains poorly defined. Genetically engineered mouse models are potentially valuable for understanding the pathogenesis of GCTs. Mice harboring constitutively active TGFß signaling (TGFBR1-CA) develop ovarian GCTs that phenocopy several hormonal and molecular characteristics of human GCTs. To determine molecular alterations in the ovary upon TGFß signaling activation, we performed transcriptomic profiling of gene expression associated with GCT development using ovaries from 1-month-old TGFBR1-CA mice and age-matched controls. RNA-sequencing and bioinformatics analysis coupled with the validation of select target genes revealed dysregulations of multiple cellular events and signaling molecules/pathways. The differentially expressed genes are enriched not only for known GCT-related pathways and tumorigenic events but also for signaling events potentially mediated by neuroactive ligand-receptor interaction, relaxin signaling, insulin signaling, and complements in TGFBR1-CA ovaries. Additionally, a comparative analysis of our data in mice with genes dysregulated in human GCTs or granulosa cells overexpressing a mutant FOXL2, the genetic hallmark of adult GCTs, identified some common genes altered in both conditions. In summary, this study has revealed the molecular signature of ovarian GCTs in a mouse model that harbors the constitutive activation of TGFBR1. The findings may be further exploited to understand the pathogenesis of a class of poorly defined ovarian tumors.

8.
J Cachexia Sarcopenia Muscle ; 13(3): 1686-1703, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35277933

RESUMEN

BACKGROUND: Cancer patients at advanced stages experience a severe depletion of skeletal muscle compartment together with a decrease in muscle function, known as cancer cachexia. Cachexia contributes to reducing quality of life, treatment efficiency, and lifespan of cancer patients. However, the systemic nature of the syndrome is poorly documented. Here, we hypothesize that glucocorticoids would be important systemic mediators of cancer cachexia. METHODS: To explore the role of glucocorticoids during cancer cachexia, biomolecular analyses were performed on several tissues (adrenal glands, blood, hypothalamus, liver, and skeletal muscle) collected from ApcMin/+ male mice, a mouse model of intestine and colon cancer, aged of 13 and 23 weeks, and compared with wild type age-matched C57BL/6J littermates. RESULTS: Twenty-three-week-old Apc mice recapitulated important features of cancer cachexia including body weight loss (-16%, P < 0.0001), muscle atrophy (gastrocnemius muscle: -53%, P < 0.0001), and weakness (-50% in tibialis anterior muscle force, P < 0.0001), increased expression of atrogens (7-fold increase in MuRF1 transcript level, P < 0.0001) and down-regulation of Akt-mTOR pathway (3.3-fold increase in 4EBP1 protein content, P < 0.0001), together with a marked transcriptional rewiring of hepatic metabolism toward an increased expression of gluconeogenic genes (Pcx: +90%, Pck1: +85%), and decreased expression of glycolytic (Slc2a2: -40%, Gk: -30%, Pklr: -60%), ketogenic (Hmgcs2: -55%, Bdh1: -80%), lipolytic/fatty oxidation (Lipe: -50%, Mgll: -60%, Cpt2: -60%, Hadh: -30%), and lipogenic (Acly: -30%, Acacb: -70%, Fasn: -45%) genes. The hypothalamic pituitary-adrenal axis was activated, as evidenced by the increase in the transcript levels of genes encoding corticotropin-releasing hormone in the hypothalamus (2-fold increase, P < 0.01), adrenocorticotropic hormone receptor (3.4-fold increase, P < 0.001), and steroid biosynthesis enzymes (Cyp21a1, P < 0.0001, and Cyp11b1, P < 0.01) in the adrenal glands, as well as by the increase in corticosterone level in the serum (+73%, P < 0.05), skeletal muscle (+17%, P < 0.001), and liver (+24%, P < 0.05) of cachectic 23-week-old Apc mice. A comparative transcriptional analysis with dexamethasone-treated C57BL/6J mice indicated that the activation of the hypothalamic-pituitary-adrenal axis in 23-week-old ApcMin/+ mice was significantly associated with the transcription of glucocorticoid-responsive genes in skeletal muscle (P < 0.05) and liver (P < 0.001). The transcriptional regulation of glucocorticoid-responsive genes was also observed in the gastrocnemius muscle of Lewis lung carcinoma tumour-bearing mice and in KPC mice (tibialis anterior muscle and liver). CONCLUSIONS: These findings highlight the role of the hypothalamic-pituitary-adrenal-glucocorticoid pathway in the transcriptional regulation of skeletal muscle catabolism and hepatic metabolism during cancer cachexia. They also provide the paradigm for the design of new therapeutic strategies.


Asunto(s)
Carcinoma Pulmonar de Lewis , Sistema Hipófiso-Suprarrenal , Anciano , Animales , Caquexia/genética , Caquexia/metabolismo , Carcinoma Pulmonar de Lewis/patología , Expresión Génica , Glucocorticoides , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/patología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/patología , Calidad de Vida
9.
Nat Immunol ; 22(9): 1140-1151, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34426691

RESUMEN

Tissue-resident memory T (TRM) cells are non-recirculating cells that exist throughout the body. Although TRM cells in various organs rely on common transcriptional networks to establish tissue residency, location-specific factors adapt these cells to their tissue of lodgment. Here we analyze TRM cell heterogeneity between organs and find that the different environments in which these cells differentiate dictate TRM cell function, durability and malleability. We find that unequal responsiveness to TGFß is a major driver of this diversity. Notably, dampened TGFß signaling results in CD103- TRM cells with increased proliferative potential, enhanced function and reduced longevity compared with their TGFß-responsive CD103+ TRM counterparts. Furthermore, whereas CD103- TRM cells readily modified their phenotype upon relocation, CD103+ TRM cells were comparatively resistant to transdifferentiation. Thus, despite common requirements for TRM cell development, tissue adaptation of these cells confers discrete functional properties such that TRM cells exist along a spectrum of differentiation potential that is governed by their local tissue microenvironment.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Plasticidad de la Célula/inmunología , Microambiente Celular/inmunología , Memoria Inmunológica/inmunología , Animales , Antígenos CD/inmunología , Linfocitos T CD8-positivos/citología , Femenino , Cadenas alfa de Integrinas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta1/metabolismo
10.
Immunity ; 54(8): 1698-1714.e5, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34233154

RESUMEN

Antigen-specific CD8+ T cells in chronic viral infections and tumors functionally deteriorate, a process known as exhaustion. Exhausted T cells are sustained by precursors of exhausted (Tpex) cells that self-renew while continuously generating exhausted effector (Tex) cells. However, it remains unknown how Tpex cells maintain their functionality. Here, we demonstrate that Tpex cells sustained mitochondrial fitness, including high spare respiratory capacity, while Tex cells deteriorated metabolically over time. Tpex cells showed early suppression of mTOR kinase signaling but retained the ability to activate this pathway in response to antigen receptor signals. Early transient mTOR inhibition improved long-term T cell responses and checkpoint inhibition. Transforming growth factor-ß repressed mTOR signaling in exhausted T cells and was a critical determinant of Tpex cell metabolism and function. Overall, we demonstrate that the preservation of cellular metabolism allows Tpex cells to retain long-term functionality to sustain T cell responses during chronic infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Metabolismo Energético/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Transducción de Señal/inmunología
12.
Immunity ; 54(1): 84-98.e5, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33212014

RESUMEN

Following antigen-driven expansion in lymph node, transforming growth factor-ß (TGFß) is required for differentiation of skin-recruited CD8+ T cell effectors into epidermal resident memory T (Trm) cells and their epidermal persistence. We found that the source of TGFß -supporting Trm cells was autocrine. In addition, antigen-specific Trm cells that encountered cognate antigen in the skin, and bystander Trm cells that did not, both displayed long-term persistence in the epidermis under steady-state conditions. However, when the active-TGFß was limited or when new T cell clones were recruited into the epidermis, antigen-specific Trm cells were more efficiently retained than bystander Trm cells. Genetically enforced TGFßR signaling allowed bystander Trm cells to persist in the epidermis as efficiently as antigen-specific Trm cells in both contexts. Thus, competition between T cells for active TGFß represents an unappreciated selective pressure that promotes the accumulation and persistence of antigen-specific Trm cells in the epidermal niche.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epidermis/inmunología , Queratinocitos/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Unión Competitiva , Efecto Espectador , Microambiente Celular , Células Clonales , Memoria Inmunológica , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Transducción de Señal , Especificidad del Receptor de Antígeno de Linfocitos T
13.
Sci Rep ; 10(1): 3880, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32127548

RESUMEN

Transforming growth factor (TGFß) is a secreted factor, which accumulates in tissues during many physio- and pathological processes such as embryonic development, wound healing, fibrosis and cancer. In order to analyze the effects of increased microenvironmental TGFß concentration in vivo, we developed a conditional transgenic mouse model (Flpo/Frt system) expressing bioactive TGFß in fibroblasts, a cell population present in the microenvironment of almost all tissues. To achieve this, we created the genetically-engineered [Fsp1-Flpo; FSFTGFßCA] mouse model. The Fsp1-Flpo allele consists in the Flpo recombinase under the control of the Fsp1 (fibroblast-specific promoter 1) promoter. The FSFTGFßCA allele consists in a transgene encoding a constitutively active mutant form of TGFß (TGFßCA) under the control of a Frt-STOP-Frt (FSF) cassette. The FSFTGFßCA allele was created to generate this model, and functionally validated by in vitro, ex vivo and in vivo techniques. [Fsp1-Flpo; FSFTGFßCA] animals do not present any obvious phenotype despite the correct expression of TGFßCA transgene in fibroblasts. This [Fsp1-Flpo; FSFTGFßCA] model is highly pertinent for future studies on the effect of increased microenvironmental bioactive TGFß concentrations in mice bearing Cre-dependent genetic alterations in other compartments (epithelial or immune compartments for instance). These dual recombinase system (DRS) approaches will enable scientists to study uncoupled spatiotemporal regulation of different genetic alterations within the same mouse, thus better replicating the complexity of human diseases.


Asunto(s)
Fibroblastos/metabolismo , Factor de Crecimiento Transformador beta/genética , Animales , Expresión Génica , Ingeniería Genética , Células Hep G2 , Humanos , Ratones , Ratones Transgénicos , Modelos Animales
14.
Genesis ; 58(5): e23359, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32191380

RESUMEN

Recombination systems represent a major breakthrough in the field of genetic model engineering. The Flp recombinases (Flp, Flpe, and Flpo) bind and cleave DNA Frt sites. We created a transgenic mouse strain ([Fsp1-Flpo]) expressing the Flpo recombinase in fibroblasts. This strain was obtained by random insertion inside mouse zygotes after pronuclear injection. Flpo expression was placed under the control of the promoter of Fsp1 (fibroblast-specific protein 1) gene, whose expression starts after gastrulation at Day 8.5 in cells of mesenchymal origin. We verified the correct expression and function of the Flpo enzyme by several ex vivo and in vivo approaches. The [Fsp1-Flpo] strain represents a genuine tool to further target the recombination of transgenes with Frt sites specifically in cells of mesenchymal origin or with a fibroblastic phenotype.


Asunto(s)
ADN Nucleotidiltransferasas/genética , Proteína de Unión al Calcio S100A4/genética , Animales , Células Cultivadas , ADN Nucleotidiltransferasas/metabolismo , Fibroblastos/metabolismo , Gástrula/metabolismo , Marcación de Gen/métodos , Células HaCaT , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas , Cigoto/metabolismo
16.
Cell Death Dis ; 10(12): 886, 2019 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-31767842

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the solid tumors with the poorest prognosis. The stroma of this tumor is abundant and composed of extracellular matrix and stromal cells (including cancer-associated fibroblasts and immune cells). Nerve fibers invading this stroma represent a hallmark of PDAC, involved in neural remodeling, which participates in neuropathic pain, cancer cell dissemination and tumor relapse after surgery. Pancreatic cancer-associated neural remodeling is regulated through functional interplays mediated by physical and molecular interactions between cancer cells, nerve cells and surrounding Schwann cells, and other stromal cells. In the present study, we show that Schwann cells (glial cells supporting peripheral neurons) can enhance aggressiveness (migration, invasion, tumorigenicity) of pancreatic cancer cells in a transforming growth factor beta (TGFß)-dependent manner. Indeed, we reveal that conditioned medium from Schwann cells contains high amounts of TGFß able to activate the TGFß-SMAD signaling pathway in cancer cells. We also observed in human PDAC samples that high levels of TGFß signaling activation were positively correlated with perineural invasion. Secretome analyses by mass spectrometry of Schwann cells and pancreatic cancer cells cultured alone or in combination highlighted the central role of TGFß in neuro-epithelial interactions, as illustrated by proteomic signatures related to cell adhesion and motility. Altogether, these results demonstrate that Schwann cells are a meaningful source of TGFß in PDAC, which plays a crucial role in the acquisition of aggressive properties by pancreatic cancer cells.

17.
Immunity ; 50(5): 1249-1261.e5, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30952606

RESUMEN

Regulated activation of the cytokine TGF-ß by integrins αvß6 and αvß8 expressed on keratinocytes is required for residence of epidermal-resident memory T cells, but whether skin-derived signals also affect recirculating memory cells in the skin remains unclear. Here, we show that after resolution of skin vaccinia virus (VV) infection, antigen-specific circulating memory CD8+ T cells migrated into skin. In mice lacking αvß6 and αvß8 integrins (Itgb6-/-Itgb8fl/fl-K14-cre), the absence of epidermal-activated TGF-ß resulted in a gradual loss of E- or P-selectin-binding central and peripheral memory populations, which were rescued when skin entry was inhibited. Skin recirculating memory cells were required for optimal host defense against skin VV infection. These data demonstrate that skin migration can persist after resolution of local skin infection and that the cytokine environment within this nonlymphoid tissue shapes the differentiation state and persistence of the central and peripheral memory-T-cell pool.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Integrinas/metabolismo , Queratinocitos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Virus Vaccinia/inmunología , Animales , Antígenos de Neoplasias/genética , Linfocitos T CD8-positivos/enzimología , Diferenciación Celular/inmunología , Citocinas/inmunología , Activación Enzimática , Femenino , Integrinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Piel/citología , Piel/inmunología
18.
Bull Cancer ; 106(5): 407-420, 2019 May.
Artículo en Francés | MEDLINE | ID: mdl-30987751

RESUMEN

INTRODUCTION: The demographics of oncology residents has changed since 2010 with the increase in the size of promotions. The evolution of the residents' aspirations towards research and their future exercise in parallel with these demographic changes has not been assessed. METHODS: A questionnaire was developed by a working group from GERCOR (cooperative group in oncology), involving clinicians, researchers, GERCOR members, and residents. It consisted of 62 questions divided into 7 sections: demographics, medical thesis, post-residency, mobility, publication activity, basic research, and clinical/translational research. The national survey was published online by the Association d'enseignement et de recherche des internes en oncologie (AERIO). RESULTS: In total, 143 residents participated, of which 116 (81.1%) completed the questionnaire entirely. The population was representative of the current demographics, with a majority of women (65.0%), a median age of 28 years, and 39.7% of residents from Paris region. The unsupervised analysis revealed four profiles of residents, including one group strongly committed to research (16.8%), one group with moderate involvement (41.3%) and one group that did not seem interested in research (14.7%). Uncertainty about future position and lack of time and interaction with researchers appeared to be the main barriers to involvement of residents in research. DISCUSSION: This national survey provided useful information about the residents' perspective to academic research. It may serve as a basis for proposing measures adapted to their expectations.


Asunto(s)
Investigación Biomédica , Internado y Residencia , Oncología Médica/educación , Francia , Autoinforme
19.
PLoS One ; 13(12): e0209417, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30550590

RESUMEN

Uterine gland development, also known as adenogenesis, is a key uterine morphogenic process indispensable for normal uterine function and fertility. Our earlier studies have reported that overactivation of TGFB receptor 1 (TGFBR1) in the mouse uterus using progesterone receptor (Pgr)-Cre recombinase causes female infertility, defective decidualization, and reduced uterine gland formation, a developmental milestone of postnatal uterus. To understand mechanisms that underpin the disrupted uterine gland formation in mice with sustained activation of TGFBR1, we raised the question of whether early postnatal adenogenesis was compromised in these mice. Experiments were designed using mice with constitutive activation of TGFBR1 driven by Pgr-Cre to determine the timing of adenogenic defects and potential mechanisms associated with dysregulation of adenogenic genes, luminal epithelial cell proliferation and endometrial fibrotic changes. Uterine tissues from mice with constitutive activation of TGFBR1 were collected during the critical time window of adenogenesis and analyzed together with age-matched controls. Multiple approaches including immunohistochemistry, immunofluorescence, Trichrome staining, quantitative real-time PCR, western blot, conditional knockout and human endometrial cell culture were utilized. TGFBR1 activation in the mouse uterus suppressed adenogenesis during postnatal uterine development, concomitant with the aberrant differentiation of uterine stromal cells. Analysis of transcript expression of WNT pathway components revealed dysregulation of adenogenesis-associated genes. Notably, the adenogenic defects occurred in spite of the increased proliferation of uterine luminal epithelial cells, accompanied by increased expression of genes associated with fibrotic changes. Moreover, the adenogenic defects were alleviated in mice where TGFBR1 was activated in presumably half of the complement of uterine cells. Our results suggest that altered differentiation of endometrial stromal cells and formation of stromal compartment promote adenogenic defects.


Asunto(s)
Células Epiteliales/fisiología , Infertilidad Femenina/etiología , Transducción de Señal/fisiología , Células del Estroma/fisiología , Útero/embriología , Animales , Diferenciación Celular , Línea Celular , Femenino , Humanos , Ratones , Ratones Transgénicos , Organogénesis , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Receptores de Progesterona/genética , Factor de Crecimiento Transformador beta/metabolismo , Útero/citología , Útero/fisiología
20.
Dig Liver Dis ; 50(10): 979-990, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30205952

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is the digestive cancer with the poorest prognosis, with a 5-year overall survival rate of 7%. Complete surgical resection followed by adjuvant chemotherapy is the only treatment with curative intent. However, many patients with an apparently localized disease who may undergo primary tumor resection already have micro-metastatic disease and will promptly develop metastases. Considering the significant rate of morbidity and mortality upon pancreatic surgery, the pre-operative identification of patients with an aggressive disease is therefore a major clinical issue. Although tumor size, differentiation, margins, and lymph node invasion are the main "classical" prognostic factors, they are not sufficient to fully predict early disease recurrence. In the last decade, multi-omics high-throughput analyses have provided a new insight into PDAC biology and have led to the description of multiple molecular subtypes, with a significant prognostic value for most of them, but that have not yet been transposed to routine clinical practice, mainly due to poor availability of tumor tissue material prior to surgical resection. In this review, we provide an overview of the current status of clinico-pathological and molecular biomarkers (tumor and blood) to predict early recurrence, and their implications for clinical practice and future research development.


Asunto(s)
Biomarcadores/sangre , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/terapia , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/terapia , Carcinoma Ductal Pancreático/mortalidad , Quimioterapia Adyuvante , Humanos , Ganglios Linfáticos/patología , Márgenes de Escisión , Clasificación del Tumor , Invasividad Neoplásica , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Pancreatectomía , Neoplasias Pancreáticas/mortalidad , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA