Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Development ; 149(19)2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36205077

RESUMEN

Notch3 promotes mammary luminal cell specification and forced Notch3 activation can induce mammary tumor formation. However, recent studies suggest a tumor-suppressive role for Notch3. Here, we report on Notch3 expression and functional analysis in the mouse mammary gland. Notch3 is expressed in the luminal compartment throughout mammary gland development, but switches to basal cells with initiation of post-lactational involution. Deletion of Notch3 caused a decrease of Notch activation in luminal cells and diminished luminal progenitors at puberty, as well as reduced alveolar progenitors during pregnancy. Parous Notch3-/- mammary glands developed hyperplasia with accumulation of CD24hiCD49flo cells, some of which progressed to invasive tumors with luminal features. Notch3 deletion abolished Notch activation in basal cells during involution, accompanied by altered apoptosis and reduced brown adipocytes, leading to expansion of parity-identified mammary epithelial cells (PI-MECs). Interestingly, the postpartum microenvironment is required for the stem cell activity of Notch3-/- PI-MECs. Finally, high expression of NOTCH3 is associated with prolonged survival in patients with luminal breast cancer. These results highlight an unexpected tumor-suppressive function for Notch3 in the parous mammary gland through restriction of PI-MEC expansion.


Asunto(s)
Células Epiteliales , Glándulas Mamarias Animales , Animales , Células Epiteliales/metabolismo , Femenino , Lactancia , Ratones , Ratones Transgénicos , Embarazo , Células Madre
2.
Mol Genet Genomics ; 299(1): 75, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39085660

RESUMEN

The clustered regularly interspaced short palindromic repeats (CRISPR) and their associated protein (Cas) system is a gene editing technology guided by RNA endonuclease. The CRISPR-Cas12a (also known as CRISPR-Cpf1) system is extensively utilized in genome editing research due to its accuracy and high efficiency. In this paper, we primarily focus on the application of CRISPR-Cpf1 technology in the construction of disease models and gene therapy. Firstly, the structure and mechanism of the CRISPR-Cas system are introduced. Secondly, the similarities and differences between CRISPR-Cpf1 and CRISPR-Cas9 technologies are compared. Thirdly, the main focus is on the application of the CRISPR-Cpf1 system in cell and animal genome editing. Finally, the challenges faced by CRISPR-Cpf1 technology and corresponding strategies are analyzed. Although CRISPR-Cpf1 technology has certain off-target effects, it can effectively and accurately edit cell and animal genomes, and has significant advantages in the preclinical research.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Edición Génica/métodos , Animales , Sistemas CRISPR-Cas/genética , Terapia Genética/métodos , Genoma/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Humanos , Proteínas Bacterianas , Endodesoxirribonucleasas , Proteínas Asociadas a CRISPR
3.
Anim Genet ; 55(1): 99-109, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38087834

RESUMEN

Niemann-Pick disease type C1 (NPC1) is a lysosomal lipid storage disease caused by NPC1 gene mutation. Our previous study found that, compared with wild-type (Npc1+/+ ) mice, the renal volume and weight of Npc1 gene mutant (Npc1-/- ) mice were significantly reduced. We speculate that Npc1 gene mutations may affect the basic structure of the kidneys of Npc1-/- mice, and thus affect their function. Therefore, we randomly selected postnatal Day 28 (P28) and P56 Npc1+/+ and Npc1-/- mice, and observed the renal structure and pathological changes by haematoxylin-eosin staining. The level of renal fibrosis was detected by immunofluorescence histochemical techniques, and western blotting was used to detect the expression levels of apoptosis-related proteins and canonical Wnt signalling pathway related proteins. The results showed that compared with Npc1+/+ mice, the kidneys of P28 and P56 Npc1-/- mice underwent apoptosis and fibrosis; furthermore, there were obvious vacuoles in the cytoplasm of renal tubular epithelial cells of P56 Npc1-/- mice, the cell bodies were loose and foam-like, and the canonical Wnt signalling pathway was abnormally activated. These results showed that Npc1 gene mutation can cause pathological changes in the kidneys of mice. As age increased, vacuoles developed in the cytoplasm of renal tubular epithelial cells, and apoptosis of renal cells, abnormal activation of the Wnt signalling pathway, and promotion of renal fibrosis increased.


Asunto(s)
Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C , Animales , Ratones , Fibrosis , Riñón/metabolismo , Riñón/patología , Mutación , Proteína Niemann-Pick C1/genética , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/metabolismo , Enfermedad de Niemann-Pick Tipo C/patología
4.
Int J Mol Sci ; 25(13)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-39000300

RESUMEN

Maize is an important crop used for food, feed, and fuel. Abiotic stress is an important factor affecting maize yield. The EPF/EPFL gene family encodes class-specific secretory proteins that play an important role in the response to abiotic stress in plants. In order to explore and utilize the EPF/EPFL family in maize, the family members were systematically identified, and their chromosomal localization, physicochemical properties, cis-acting element prediction in promoters, phylogenetic tree construction, and expression pattern analysis were carried out using bioinformatics techniques. A total of 18 ZmEPF/EPFL proteins were identified in maize, which are mostly alkaline and a small portion acidic. Subcellular localization results showed that ZmEPF6, ZmEPF12, and ZmEPFL2 are localized in the nucleus and cytoplasm. Analysis of cis-acting elements revealed that members of the ZmEPF/EPFL family contain regulatory elements such as light response, anoxic, low temperature, and hormone response regulatory elements. RT-qPCR results showed that these family members are indeed responding to cold stress and hormone treatments. These results of this study provide a theoretical basis for improving the abiotic stress resistance of maize in future research.


Asunto(s)
Regulación de la Expresión Génica de las Plantas , Familia de Multigenes , Filogenia , Proteínas de Plantas , Estrés Fisiológico , Zea mays , Zea mays/genética , Zea mays/metabolismo , Zea mays/fisiología , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Estrés Fisiológico/genética , Regiones Promotoras Genéticas
5.
EMBO J ; 38(13): e101067, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31268604

RESUMEN

A prominent function of TGIF1 is suppression of transforming growth factor beta (TGF-ß) signaling, whose inactivation is deemed instrumental to the progression of pancreatic ductal adenocarcinoma (PDAC), as exemplified by the frequent loss of the tumor suppressor gene SMAD4 in this malignancy. Surprisingly, we found that genetic inactivation of Tgif1 in the context of oncogenic Kras, KrasG12D , culminated in the development of highly aggressive and metastatic PDAC despite de-repressing TGF-ß signaling. Mechanistic experiments show that TGIF1 associates with Twist1 and inhibits Twist1 expression and activity, and this function is suppressed in the vast majority of human PDACs by KrasG12D /MAPK-mediated TGIF1 phosphorylation. Ablating Twist1 in KrasG12D ;Tgif1KO mice completely blunted PDAC formation, providing the proof-of-principle that TGIF1 restrains KrasG12D -driven PDAC through its ability to antagonize Twist1. Collectively, these findings pinpoint TGIF1 as a potential tumor suppressor in PDAC and further suggest that sustained activation of TGF-ß signaling might act to accelerate PDAC progression rather than to suppress its initiation.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Metástasis de la Neoplasia , Proteínas Nucleares/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteína 1 Relacionada con Twist/genética
6.
Am J Pathol ; 190(11): 2194-2202, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32805234

RESUMEN

The prostate epithelium consists of predominantly luminal cells that express androgen receptor and require androgens for growth. As a consequence, the depletion of testicular androgens in patients with prostate cancer results in tumor regression. However, it eventually leads to a castration-resistant disease that is highly metastatic. In this report, a mouse model of metastatic prostate cancer was generated through the deletion of the tumor-suppressor gene Trp53 in conjunction with oncogenic activation of the proto-oncogene Kras. These mice developed early-onset metastatic prostate cancer with complete penetrance. Tumors from these mice were poorly differentiated adenocarcinoma, characterized by extensive epithelial-mesenchymal transition. With no or a very low level of androgen receptor expression, the tumor cells were resistant to androgen receptor inhibition. Pik3cg, encoding phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit γ (Pi3kγ), was highly expressed in these tumors, and pharmacologic inhibition of Pi3kγ blocked tumor cell growth in vitro, reversed epithelial-mesenchymal transition, and abated tumor metastasis in vivo. Immunohistochemistry analysis in human prostate cancer specimens showed that the expression of PIK3CG was significantly associated with advanced clinical stages. Taken together, these results suggest that PIK3CG plays an important role in the progression and metastasis of prostate cancer, and may represent a new therapeutic target in the metastatic castration-resistant prostate cancer.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Animales , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Masculino , Ratones , Ratones Transgénicos , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proto-Oncogenes Mas , Receptores Androgénicos/genética
7.
PLoS Genet ; 14(9): e1007660, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30188892

RESUMEN

FGF signaling is a potent inducer of lacrimal gland development in the eye, capable of transforming the corneal epithelium into glandular tissues. Here, we show that genetic ablation of the Pea3 family of transcription factors not only disrupted the ductal elongation and branching of the lacrimal gland, but also biased the lacrimal gland epithelium toward an epidermal cell fate. Analysis of high-throughput gene expression and chromatin immunoprecipitation data revealed that the Pea3 genes directly control both the positive and negative feedback loops of FGF signaling. Importantly, Pea3 genes are also required to suppress aberrant Notch signaling which, if gone unchecked, can compromise lacrimal gland development by preventing the expression of both Sox and Six family genes. These results demonstrate that Pea3 genes are key FGF early response transcriptional factors, programing the genetic landscape for cell fate determination.


Asunto(s)
Diferenciación Celular/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Aparato Lagrimal/crecimiento & desarrollo , Factores de Transcripción/metabolismo , Animales , Células Epidérmicas/fisiología , Células Epiteliales/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Aparato Lagrimal/citología , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Receptores Notch/metabolismo , Factores de Transcripción SOX/genética , Factores de Transcripción SOX/metabolismo , Factores de Transcripción/genética
8.
Immunity ; 30(2): 254-63, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19217325

RESUMEN

Notch2 activation induced by Delta-like-1 (DL1) drives development of splenic marginal zone (MZ) B cells, an innate-like lineage that protects against sepsis. DL1 interacts with Notch2 weakly, but it is not known whether enhancement of DL1-induced Notch2 activation by Fringe glycosyltransferases is important for MZ B cell development. Furthermore, DL1-expressing cells that promote MZ B cell development have not been identified. We show that Lunatic Fringe (Lfng) and Manic Fringe (Mfng) cooperatively enhanced the DL1-Notch2 interaction to promote MZ B cell development. We also identified radio-resistant red pulp endothelial cells in the splenic MZ that express high amounts of DL1 and promoted MZ B generation. Finally, MZ B cell precursor competition for DL1 homeostatically regulated entry into the MZ B cell pool. Our study has revealed that the Fringe-Notch2 interaction has important functions in vivo and provides insights into mechanisms regulating MZ B cell development.


Asunto(s)
Linfocitos B/citología , Linfocitos B/inmunología , Células Endoteliales/inmunología , Glicosiltransferasas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas/metabolismo , Bazo/inmunología , Animales , Fusión Artificial Génica , Linfocitos B/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas de Unión al Calcio , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Células Endoteliales/citología , Glucosiltransferasas , Glicosiltransferasas/deficiencia , Glicosiltransferasas/genética , Glicosiltransferasas/inmunología , Proteínas de Homeodominio , Ratones , Ratones Noqueados , Proteínas/genética , Proteínas/inmunología , ARN Mensajero/genética , Receptor Notch2/metabolismo , Bazo/citología , Bazo/metabolismo , Factor de Transcripción HES-1
9.
Dev Dyn ; 242(6): 678-86, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23526493

RESUMEN

BACKGROUND: The Notch signaling pathway plays complex roles in developing lungs, including regulation of proximodistal fates, airway cell specification and differentiation. However, the specific Notch-mediated signals involved in lung development remain unclear. RESULTS: Here we report that Jagged1 is expressed in a subset of bronchial and bronchiolar epithelial cells, where it controls proximal airway cell fate and differentiation. In agreement with previous studies involving disruption of all Notch signaling, we found that deletion of Jagged1 in airway epithelium increased the number of ciliated cells at the expense of Clara cells, a phenotype associated with downregulation of Hes1. Deletion of Jagged1 also led to an increased number of pulmonary neuroendocrine cells (PNEC), suggesting that Jagged1/Notch signaling inhibits PNEC cell fate. As expected, Jagged1 deletion did not affect alveolar cell differentiation, although alveolar septation was impaired, likely an indirect effect of proximal airway defects. Finally, in the postnatal lung, Jagged1 deletion induced mucous metaplasia, accompanied by downregulation of Hes1 and Hes5. CONCLUSIONS: Our results demonstrate that Jagged1-mediated Notch signaling regulates multiple cell fate decisions as well as differentiation in the respiratory system to coordinate lung development and to maintain a balance of airway cell types in adult life.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/fisiología , Pulmón/embriología , Proteínas de la Membrana/fisiología , Receptores Notch/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Diferenciación Celular , Células Epiteliales/metabolismo , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/genética , Proteína Jagged-1 , Pulmón/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Células Neuroendocrinas/metabolismo , Fenotipo , Proteínas Serrate-Jagged , Transducción de Señal , Factores de Tiempo
10.
Front Oncol ; 14: 1373760, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38646436

RESUMEN

Colorectal cancer, with the liver being the most common site of distant metastasis, followed by the lungs and bones. Although reports of metastasis to the testis exist, paratesticular metastasis is extremely rare. A 37-year-old male presented with scrotal swelling. Ultrasound revealed hydrocele of the tunica vaginalis. The patient underwent routine surgical treatment, and postoperative pathology of the tunica vaginalis indicated adenocarcinoma of gastrointestinal origin. Colonoscopic biopsy confirmed adenocarcinoma of the sigmoid colon. After six months of systemic therapy, tumor reduction surgery was performed in conjunction with tunica vaginalis excision. Postoperative pathology suggested histological similarity in both sites, with immunohistochemistry results supporting the diagnosis of sigmoid colon adenocarcinoma metastasizing to the tunica vaginalis. We conducted a literature review, summarizing and discussing clinical presentations, metastatic pathways, and diagnostic approaches.

11.
Blood ; 117(4): 1184-95, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21097675

RESUMEN

Lunatic Fringe (Lfng) enhances Notch1 activation by Delta-like 4 (DL4) to promote Notch1-dependent T-lineage commitment of thymus-seeding progenitors. Subsequently, Notch1 and T-cell receptor-ß (TCRß)-containing pre-TCR complexes signal CD4/CD8 double-negative 3 (DN3) committed T-cell progenitors to survive, proliferate, and differentiate into CD4/CD8 double-positive (DP) αß T-cell precursors. Few DP thymocytes develop without Notch1 or pre-TCR signals, whereas ectopic Notch1 activation causes T-cell leukemia. However, mechanisms of a Notch-pre-TCR collaboration during this "ß-selection" process are poorly understood. We genetically manipulated Lfng to attenuate or enhance Notch1 activation in DN3 thymocytes without inducing leukemogenesis. We show that Lfng temporally sustains DL-induced Notch1 signaling to prolong proliferative self-renewal of pre-DP thymocytes. Pre-TCR signaling greatly augmented Notch trophic functions to promote robust proliferation of pre-DP progenitors. In contrast, in the absence of DL/Notch signaling, pre-TCR-expressing progenitors rapidly atrophied and differentiated into DP thymocytes. Thus, Lfng prolongs Notch1 signaling to promote self-renewal more than differentiation during the early stages of ß-selection. Our data provide novel insights into the Notch-pre-TCR collaboration, and suggest that decreasing Lfng expression during the DN3-DP transition minimizes the potent leukemogenic potential of Notch1 signaling.


Asunto(s)
Proliferación Celular , Glicosiltransferasas/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Células Progenitoras Linfoides/fisiología , Proteínas de la Membrana/fisiología , Receptor Notch1/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/fisiología , Proteínas de Unión al Calcio , Células Cultivadas , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Progenitoras Linfoides/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/metabolismo
12.
Adv Cancer Res ; 159: 1-36, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37268393

RESUMEN

The Notch signaling pathway is an evolutionary conserved signal transduction cascade that is critical to embryonic and postnatal development, but aberrant Notch signaling is also implicated in tumorigenesis of many organs including the pancreas. Pancreatic ductal adenocarcinoma (PDAC) is the most common malignancy in the pancreas, with a dismally low survival rate due to the late-stage diagnosis and peculiar therapeutic resistance. Upregulation of the Notch signaling pathway has been found in preneoplastic lesions as well as PDACs in genetically engineered mouse models and human patients, and inhibition of the Notch signaling suppresses tumor development and progression in mice as well as patient-derived xenograft tumor growth, suggesting a critical role for Notch in PDAC. However, the role of Notch signaling pathway remains contentious, exemplified by differential functions of Notch receptors and contrasting outcomes of abolishing Notch signaling in murine PDAC models with distinct cell-of-origin or at different stages. Glycosylation of Notch receptors represents a powerful regulatory mechanism of Notch signaling, and its functional significance in PDAC has begun to emerge. Beyond its impact on tumor cells, Notch signaling is an important regulator of the components of pancreatic tumor microenvironment, including blood vasculature, stellate cells, fibroblasts, and immune cells. Finally, Notch may act as a tumor suppressor in pancreatic neuroendocrine tumor, the second most common pancreatic neoplasm with the incidence on rise. This review summarizes the research on the complex roles of Notch signaling in pancreatic tumorigenesis and the development of potential Notch-targeting therapies for pancreatic cancer.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Neoplasias Pancreáticas/metabolismo , Transformación Celular Neoplásica , Carcinogénesis , Transducción de Señal , Carcinoma Ductal Pancreático/metabolismo , Páncreas/metabolismo , Páncreas/patología , Receptores Notch/metabolismo , Microambiente Tumoral , Neoplasias Pancreáticas
13.
J Cell Biol ; 222(4)2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36828547

RESUMEN

The transcription factor Prdm16 functions as a potent suppressor of transforming growth factor-beta (TGF-ß) signaling, whose inactivation is deemed essential to the progression of pancreatic ductal adenocarcinoma (PDAC). Using the KrasG12D-based mouse model of human PDAC, we surprisingly found that ablating Prdm16 did not block but instead accelerated PDAC formation and progression, suggesting that Prdm16 might function as a tumor suppressor in this malignancy. Subsequent genetic experiments showed that ablating Prdm16 along with Smad4 resulted in a shift from a well-differentiated and confined neoplasm to a highly aggressive and metastatic disease, which was associated with a striking deviation in the trajectory of the premalignant lesions. Mechanistically, we found that Smad4 interacted with and recruited Prdm16 to repress its own expression, therefore pinpointing a model in which Prdm16 functions downstream of Smad4 to constrain the PDAC malignant phenotype. Collectively, these findings unveil an unprecedented antagonistic interaction between the tumor suppressors Smad4 and Prdm16 that functions to restrict PDAC progression and metastasis.


Asunto(s)
Carcinoma Ductal Pancreático , Proteínas de Unión al ADN , Neoplasias Pancreáticas , Proteína Smad4 , Factores de Transcripción , Animales , Humanos , Ratones , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteína Smad4/genética , Proteína Smad4/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neoplasias Pancreáticas
14.
Front Immunol ; 14: 1244159, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37901240

RESUMEN

Introduction: Triple-negative breast cancer (TNBC) comprises a heterogeneous group of clinically aggressive tumors with high risk of recurrence and metastasis. Current pharmacological treatment options remain largely limited to chemotherapy. Despite promising results, the efficacy of immunotherapy and chemo-immunotherapy in TNBC remains limited. There is strong evidence supporting the involvement of Notch signaling in TNBC progression. Expression of Notch1 and its ligand Jagged1 correlate with poor prognosis. Notch inhibitors, including g-secretase inhibitors (GSIs), are quite effective in preclinical models of TNBC. However, the success of GSIs in clinical trials has been limited by their intestinal toxicity and potential for adverse immunological effects, since Notch plays key roles in T-cell activation, including CD8 T-cells in tumors. Our overarching goal is to replace GSIs with agents that lack their systemic toxicity and ideally, do not affect tumor immunity. We identified sulindac sulfide (SS), the active metabolite of FDA-approved NSAID sulindac, as a potential candidate to replace GSIs. Methods: We investigated the pharmacological and immunotherapeutic properties of SS in TNBC models in vitro, ex-vivo and in vivo. Results: We confirmed that SS, a known γ-secretase modulator (GSM), inhibits Notch1 cleavage in TNBC cells. SS significantly inhibited mammosphere growth in all human and murine TNBC models tested. In a transplantable mouse TNBC tumor model (C0321), SS had remarkable single-agent anti-tumor activity and eliminated Notch1 protein expression in tumors. Importantly, SS did not inhibit Notch cleavage in T- cells, and the anti-tumor effects of SS were significantly enhanced when combined with a-PD1 immunotherapy in our TNBC organoids and in vivo. Discussion: Our data support further investigation of SS for the treatment of TNBC, in conjunction with chemo- or -chemo-immunotherapy. Repurposing an FDA-approved, safe agent for the treatment of TNBC may be a cost-effective, rapidly deployable therapeutic option for a patient population in need of more effective therapies.


Asunto(s)
Sulindac , Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Sulindac/farmacología , Sulindac/uso terapéutico , Secretasas de la Proteína Precursora del Amiloide , Neoplasias de la Mama Triple Negativas/metabolismo , Antiinflamatorios no Esteroideos/uso terapéutico , Modelos Animales de Enfermedad
15.
Adv Exp Med Biol ; 727: 89-98, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22399341

RESUMEN

Notch signaling plays an essential role in development and homeostasis of multiple organs including the lung. Dysregulation of Notch signaling has been implicated in various lung diseases including lung cancer. Here we review functions of Notch signaling in coordinating events during lung development, such as early proximodistal fate generation and branching, airway epithelial cell fate specification, alveogenesis and pulmonary vascular development. We also discuss roles of Notch in chronic obstructive pulmonary disease, progressive pulmonary fibrosis, pulmonary arterial hypertension, asthma and lung cancer.


Asunto(s)
Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Pulmón/citología , Pulmón/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Humanos
16.
Cell Rep ; 41(6): 111623, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36351408

RESUMEN

A long-standing question in the pancreatic ductal adenocarcinoma (PDAC) field has been whether alternative genetic alterations could substitute for oncogenic KRAS mutations in initiating malignancy. Here, we report that Neurofibromin1 (NF1) inactivation can bypass the requirement of mutant KRAS for PDAC pathogenesis. An in-depth analysis of PDAC databases reveals various genetic alterations in the NF1 locus, including nonsense mutations, which occur predominantly in tumors with wild-type KRAS. Genetic experiments demonstrate that NF1 ablation culminates in acinar-to-ductal metaplasia, an early step in PDAC. Furthermore, NF1 haploinsufficiency results in a dramatic acceleration of KrasG12D-driven PDAC. Finally, we show an association between NF1 and p53 that is orchestrated by PML, and mosaic analysis with double markers demonstrates that concomitant inactivation of NF1 and Trp53 is sufficient to trigger full-blown PDAC. Together, these findings open up an exploratory framework for apprehending the mechanistic paradigms of PDAC with normal KRAS, for which no effective therapy is available.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Mutación , Conductos Pancreáticos/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Neurofibromina 1/metabolismo , Neoplasias Pancreáticas
17.
Front Immunol ; 13: 987298, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36090975

RESUMEN

A critical feature of cancer is the ability to induce immunosuppression and evade immune responses. Tumor-induced immunosuppression diminishes the effectiveness of endogenous immune responses and decreases the efficacy of cancer immunotherapy. In this study, we describe a new immunosuppressive pathway in which adenosine promotes Casitas B-lineage lymphoma b (Cbl-b)-mediated Notch1 degradation, causing suppression of CD8+ T-cells effector functions. Genetic knockout and pharmacological inhibition of Cbl-b prevents Notch1 degradation in response to adenosine and reactivates its signaling. Reactivation of Notch1 results in enhanced CD8+ T-cell effector functions, anti-cancer response and resistance to immunosuppression. Our work provides evidence that targeting the Cbl-b-Notch1 axis is a novel promising strategy for cancer immunotherapy.


Asunto(s)
Linfoma , Neoplasias , Adenosina , Linfocitos T CD8-positivos , Humanos , Inmunoterapia , Receptor Notch1/genética , Receptor Notch1/metabolismo
18.
J Cell Physiol ; 226(7): 1940-52, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21506125

RESUMEN

Notch genes play a critical role in mammary gland growth, development and tumorigenesis. In the present study, we have quantitatively determined the levels and mRNA expression patterns of the Notch receptor genes, their ligands and target genes in the postnatal mouse mammary gland. The steady state levels of Notch3 mRNA are the highest among receptor genes, Jagged1 and Dll3 mRNA levels are the highest among ligand genes and Hey2 mRNA levels are highest among expressed Hes/Hey target genes analyzed during different stages of postnatal mammary gland development. Using an immunohistochemical approach with antibodies specific for each Notch receptor, we show that Notch proteins are temporally regulated in mammary epithelial cells during normal mammary gland development in the FVB/N mouse. The loss of ovarian hormones is associated with changes in the levels of Notch receptor mRNAs (Notch2 higher and Notch3 lower) and ligand mRNAs (Dll1 and Dll4 are higher, whereas Dll3 and Jagged1 are lower) in the mammary gland of ovariectomized mice compared to intact mice. These data define expression of the Notch ligand/receptor system throughout development of the mouse mammary gland and help set the stage for genetic analysis of Notch in this context.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Glándulas Mamarias Animales/metabolismo , Receptores Notch/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Unión al Calcio/genética , Línea Celular , Femenino , Hormonas Esteroides Gonadales/metabolismo , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína Jagged-1 , Lactancia/genética , Ligandos , Glándulas Mamarias Animales/crecimiento & desarrollo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Ovariectomía , Embarazo , Interferencia de ARN , ARN Mensajero/metabolismo , Receptor Notch2/genética , Receptor Notch3 , Receptores Notch/metabolismo , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Serrate-Jagged , Transfección
19.
Life Sci Alliance ; 4(2)2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33268505

RESUMEN

Notch signaling exerts both oncogenic and tumor-suppressive functions in the pancreas. In this study, deletion of Jag1 in conjunction with oncogenic Kras G12D expression in the mouse pancreas induced rapid development of acinar-to-ductal metaplasia and early stage pancreatic intraepithelial neoplasm; however, culminating in cystic neoplasms rather than ductal adenocarcinoma. Most cystic lesions in these mice were reminiscent of serous cystic neoplasm, and the rest resembled intraductal papillary mucinous neoplasm. Jag1 expression was lost or decreased in cystic lesions but retained in adenocarcinoma in these mice, so was the expression of Sox9. In pancreatic cancer patients, JAG1 expression is higher in cancerous tissue, and high JAG1 is associated with poor overall survival. Expression of SOX9 is correlated with JAG1, and high SOX9 is also associated with poor survival. Mechanistically, Jag1 regulates expression of Lkb1, a tumor suppressor involved in the development of pancreatic cystic neoplasm. Collectively, Jag1 can act as a tumor suppressor in the pancreas by delaying precursor lesions, whereas loss of Jag1 promoted a phenotypic switch from malignant carcinoma to benign cystic lesions.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteína Jagged-1/deficiencia , Neoplasias Pancreáticas/etiología , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Expresión Génica , Humanos , Inmunohistoquímica , Proteína Jagged-1/metabolismo , Ratones , Ratones Noqueados , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Fenotipo , Pronóstico , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factor de Transcripción SOX9 , Transducción de Señal
20.
Traffic ; 9(5): 742-54, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18298590

RESUMEN

Intersectins (Itsn) are conserved EH and SH3 domain containing adaptor proteins. In Drosophila melanogaster, ITSN is required to regulate synaptic morphology, to facilitate efficient synaptic vesicle recycling and for viability. Here, we report our genetic analysis of Caenorhabditis elegans intersectin. In contrast to Drosophila, C. elegans itsn-1 protein null mutants are viable and display grossly normal locomotion and development. However, motor neurons in these mutants show a dramatic increase in large irregular vesicles and accumulate membrane-associated vesicles at putative endocytic hotspots, approximately 300 nm from the presynaptic density. This defect occurs precisely where endogenous ITSN-1 protein localizes in wild-type animals and is associated with a significant reduction in synaptic vesicle number and reduced frequency of endogenous synaptic events at neuromuscular junctions (NMJs). ITSN-1 forms a stable complex with EHS-1 (Eps15) and is expressed at reduced levels in ehs-1 mutants. Thus, ITSN-1 together with EHS-1, coordinate vesicle recycling at C. elegans NMJs. We also found that both itsn-1 and ehs-1 mutants show poor viability and growth in a Disabled (dab-1) null mutant background. These results show for the first time that intersectin and Eps15 proteins function in the same genetic pathway, and appear to function synergistically with the clathrin-coat-associated sorting protein, Disabled, for viability.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Unión Neuromuscular/fisiología , Vesículas Sinápticas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/anatomía & histología , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/genética , Drosophila melanogaster , Endocitosis , Eliminación de Gen , Genes Reporteros , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Técnicas de Placa-Clamp , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura , Transmisión Sináptica/fisiología , Vesículas Sinápticas/ultraestructura , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA