Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.866
Filtrar
Más filtros

Intervalo de año de publicación
1.
Immunity ; 50(4): 924-940, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30995507

RESUMEN

Transforming growth factor (TGF)-ß is a crucial enforcer of immune homeostasis and tolerance, inhibiting the expansion and function of many components of the immune system. Perturbations in TGF-ß signaling underlie inflammatory diseases and promote tumor emergence. TGF-ß is also central to immune suppression within the tumor microenvironment, and recent studies have revealed roles in tumor immune evasion and poor responses to cancer immunotherapy. Here, we present an overview of the complex biology of the TGF-ß family and its context-dependent nature. Then, focusing on cancer, we discuss the roles of TGF-ß signaling in distinct immune cell types and how this knowledge is being leveraged to unleash the immune system against the tumor.


Asunto(s)
Neoplasias/inmunología , Factor de Crecimiento Transformador beta/fisiología , Inmunidad Adaptativa , Animales , Células Dendríticas/inmunología , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Fibroblastos/inmunología , Humanos , Inmunidad Innata , Inflamación , Macrófagos/inmunología , Ratones Noqueados , Neutrófilos/inmunología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/inmunología , Escape del Tumor , Microambiente Tumoral
2.
Annu Rev Physiol ; 85: 269-291, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36266260

RESUMEN

Myostatin (GDF-8) was discovered 25 years ago as a new transforming growth factor-ß family member that acts as a master regulator of skeletal muscle mass. Myostatin is made by skeletal myofibers, circulates in the blood, and acts back on myofibers to limit growth. Myostatin appears to have all of the salient properties of a chalone, which is a term proposed over a half century ago to describe hypothetical circulating, tissue-specific growth inhibitors that control tissue size. The elucidation of the molecular, cellular, and physiological mechanisms underlying myostatin activity suggests that myostatin functions as a negative feedback regulator of muscle mass and raises the question as to whether this type of chalone mechanism is unique to skeletal muscle or whether it also operates in other tissues.


Asunto(s)
Chalonas , Miostatina , Humanos , Músculo Esquelético/fisiología , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta/fisiología
3.
Immunity ; 47(5): 903-912.e4, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29126797

RESUMEN

Alveolar macrophages (AMs) derive from fetal liver monocytes, which colonize the lung during embryonic development and give rise to fully mature AMs perinatally. AM differentiation requires granulocyte macrophage colony-stimulating factor (GM-CSF), but whether additional factors are involved in AM regulation is not known. Here we report that AMs, in contrast to most other tissue macrophages, were also dependent on transforming growth factor-ß receptor (TGF-ßR) signaling. Conditional deletion of TGF-ßR in mice at different time points halted the development and differentiation of AMs. In adult mice, TGF-ß was also critical for AM homeostasis. The source of TGF-ß was AMs themselves, indicative of an autocrine loop that promotes AM self-maintenance. Mechanistically, TGF-ßR signaling resulted in upregulation of PPAR-γ, a signature transcription factor essential for the development of AMs. These findings reveal an additional layer of complexity regarding the guidance cues, which govern the genesis, maturation, and survival of AMs.


Asunto(s)
Homeostasis , Macrófagos Alveolares/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Diferenciación Celular , Desarrollo Embrionario , Ratones , Ratones Endogámicos C57BL , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal/fisiología , Transcriptoma
4.
Immunity ; 47(5): 959-973.e9, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29150241

RESUMEN

Aortic aneurysms are life-threatening conditions with effective treatments mainly limited to emergency surgery or trans-arterial endovascular stent grafts, thus calling for the identification of specific molecular targets. Genetic studies have highlighted controversial roles of transforming growth factor ß (TGF-ß) signaling in aneurysm development. Here, we report on aneurysms developing in adult mice after smooth muscle cell (SMC)-specific inactivation of Smad4, an intracellular transducer of TGF-ß. The results revealed that Smad4 inhibition activated interleukin-1ß (IL-1ß) in SMCs. This danger signal later recruited innate immunity in the adventitia through chemokine (C-C motif) ligand 2 (CCL2) and modified the mechanical properties of the aortic wall, thus favoring vessel dilation. SMC-specific Smad4 deletion in Il1r1- or Ccr2-null mice resulted in milder aortic pathology. A chronic treatment with anti-IL-1ß antibody effectively hampered aneurysm development. These findings identify a mechanistic target for controlling the progression of aneurysms with compromised TGF-ß signaling, such as those driven by SMAD4 mutations.


Asunto(s)
Aneurisma de la Aorta/prevención & control , Interleucina-1beta/antagonistas & inhibidores , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Células Cultivadas , Quimiocina CCL2/antagonistas & inhibidores , Interleucina-1beta/biosíntesis , Ratones , Miocitos del Músculo Liso/inmunología , FN-kappa B/fisiología , Receptores CCR2/antagonistas & inhibidores , Proteína Smad4/fisiología , Tamoxifeno/farmacología
5.
PLoS Genet ; 17(7): e1009678, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34260587

RESUMEN

Animals can adapt to dynamic environmental conditions by modulating their developmental programs. Understanding the genetic architecture and molecular mechanisms underlying developmental plasticity in response to changing environments is an important and emerging area of research. Here, we show a novel role of cAMP response element binding protein (CREB)-encoding crh-1 gene in developmental polyphenism of C. elegans. Under conditions that promote normal development in wild-type animals, crh-1 mutants inappropriately form transient pre-dauer (L2d) larvae and express the L2d marker gene. L2d formation in crh-1 mutants is specifically induced by the ascaroside pheromone ascr#5 (asc-ωC3; C3), and crh-1 functions autonomously in the ascr#5-sensing ASI neurons to inhibit L2d formation. Moreover, we find that CRH-1 directly binds upstream of the daf-7 TGF-ß locus and promotes its expression in the ASI neurons. Taken together, these results provide new insight into how animals alter their developmental programs in response to environmental changes.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adaptación Fisiológica/genética , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Ciclo Celular , Procesos de Crecimiento Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/genética , Larva/genética , Larva/crecimiento & desarrollo , Feromonas/metabolismo , Células Receptoras Sensoriales/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/fisiología
6.
Nat Immunol ; 12(10): 1002-9, 2011 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-21874024

RESUMEN

T cell activation is subject to tight regulation to avoid inappropriate responses to self antigens. Here we show that genetic deficiency in the ubiquitin ligase Peli1 caused hyperactivation of T cells and rendered T cells refractory to suppression by regulatory T cells and transforming growth factor-ß (TGF-ß). As a result, Peli1-deficient mice spontaneously developed autoimmunity characterized by multiorgan inflammation and autoantibody production. Peli1 deficiency resulted in the nuclear accumulation of c-Rel, a member of the NF-κB family of transcription factors with pivotal roles in T cell activation. Peli1 negatively regulated c-Rel by mediating its Lys48 (K48) ubiquitination. Our results identify Peli1 as a critical factor in the maintenance of peripheral T cell tolerance and demonstrate a previously unknown mechanism of c-Rel regulation.


Asunto(s)
Autoinmunidad , Activación de Linfocitos , Proteínas Nucleares/fisiología , Linfocitos T/inmunología , Animales , Antígenos CD28/fisiología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-rel/metabolismo , Receptores de Antígenos de Linfocitos T/fisiología , Linfocitos T Reguladores/fisiología , Factor de Crecimiento Transformador beta/fisiología , Ubiquitina-Proteína Ligasas , Ubiquitinación
7.
Mol Psychiatry ; 27(1): 296-306, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34131268

RESUMEN

Neuropsychiatric diseases are manifested by maladaptive behavioral plasticity. Despite the greater understanding of the neuroplasticity underlying behavioral adaptations, pinpointing precise cellular mediators has remained elusive. This has stymied the development of pharmacological interventions to combat these disorders both at the level of progression and relapse. With increased knowledge on the putative role of the transforming growth factor (TGF- ß) family of proteins in mediating diverse neuroadaptations, the influence of TGF-ß signaling in regulating maladaptive cellular and behavioral plasticity underlying neuropsychiatric disorders is being increasingly elucidated. The current review is focused on what is currently known about the TGF-ß signaling in the central nervous system in mediating cellular and behavioral plasticity related to neuropsychiatric manifestations.


Asunto(s)
Trastornos Mentales , Enfermedades del Sistema Nervioso , Transducción de Señal , Factor de Crecimiento Transformador beta , Sistema Nervioso Central , Humanos , Plasticidad Neuronal , Factor de Crecimiento Transformador beta/fisiología
8.
PLoS Biol ; 18(9): e3000825, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32886690

RESUMEN

Microbial dysbiosis in the upper digestive tract is linked to an increased risk of esophageal squamous cell carcinoma (ESCC). Overabundance of Porphyromonas gingivalis is associated with shorter survival of ESCC patients. We investigated the molecular mechanisms driving aggressive progression of ESCC by P. gingivalis. Intracellular invasion of P. gingivalis potentiated proliferation, migration, invasion, and metastasis abilities of ESCC cells via transforming growth factor-ß (TGFß)-dependent Drosophila mothers against decapentaplegic homologs (Smads)/Yes-associated protein (YAP)/Transcriptional coactivator with PDZ-binding motif (TAZ) activation. Smads/YAP/TAZ/TEA domain transcription factor1 (TEAD1) complex formation was essential to initiate downstream target gene expression, inducing an epithelial-mesenchymal transition (EMT) and stemness features. Furthermore, P. gingivalis augmented secretion and bioactivity of TGFß through glycoprotein A repetitions predominant (GARP) up-regulation. Accordingly, disruption of either the GARP/TGFß axis or its activated Smads/YAP/TAZ complex abrogated the tumor-promoting role of P. gingivalis. P. gingivalis signature genes based on its activated effector molecules can efficiently distinguish ESCC patients into low- and high-risk groups. Targeting P. gingivalis or its activated effectors may provide novel insights into clinical management of ESCC.


Asunto(s)
Infecciones por Bacteroidaceae/complicaciones , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , Porphyromonas gingivalis/fisiología , Factor de Crecimiento Transformador beta/fisiología , Aciltransferasas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Anciano , Animales , Infecciones por Bacteroidaceae/metabolismo , Infecciones por Bacteroidaceae/mortalidad , Infecciones por Bacteroidaceae/patología , Células Cultivadas , Progresión de la Enfermedad , Drosophila , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/microbiología , Neoplasias Esofágicas/mortalidad , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/microbiología , Carcinoma de Células Escamosas de Esófago/mortalidad , Femenino , Estudios de Seguimiento , Células HCT116 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Transducción de Señal/fisiología , Proteínas Smad/metabolismo , Análisis de Supervivencia , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Señalizadoras YAP
9.
Nat Rev Mol Cell Biol ; 12(11): 722-34, 2011 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-21952300

RESUMEN

Adipose tissue, which is primarily composed of adipocytes, is crucial for maintaining energy and metabolic homeostasis. Adipogenesis is thought to occur in two stages: commitment of mesenchymal stem cells to a preadipocyte fate and terminal differentiation. Cell shape and extracellular matrix remodelling have recently been found to regulate preadipocyte commitment and competency by modulating WNT and RHO-family GTPase signalling cascades. Adipogenic stimuli induce terminal differentiation in committed preadipocytes through the epigenomic activation of peroxisome proliferator-activated receptor-γ (PPARγ). The coordination of PPARγ with CCAAT/enhancer-binding protein (C/EBP) transcription factors maintains adipocyte gene expression. Improving our understanding of these mechanisms may allow us to identify therapeutic targets against metabolic diseases that are rapidly becoming epidemic globally.


Asunto(s)
Adipocitos/citología , Adipocitos/fisiología , Adipogénesis/fisiología , Adipogénesis/genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Linaje de la Célula , Forma de la Célula , Epigénesis Genética , Matriz Extracelular/fisiología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/fisiología , Modelos Biológicos , PPAR gamma/genética , PPAR gamma/fisiología , Transducción de Señal , Factor de Crecimiento Transformador beta/fisiología , Vía de Señalización Wnt , Proteínas de Unión al GTP rho/fisiología
10.
Nucleic Acids Res ; 49(1): 127-144, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33305319

RESUMEN

MicroRNAs are regulators of gene expression. A wide-spread, yet not validated, assumption is that the targetome of miRNAs is non-randomly distributed across the transcriptome and that targets share functional pathways. We developed a computational and experimental strategy termed high-throughput miRNA interaction reporter assay (HiTmIR) to facilitate the validation of target pathways. First, targets and target pathways are predicted and prioritized by computational means to increase the specificity and positive predictive value. Second, the novel webtool miRTaH facilitates guided designs of reporter assay constructs at scale. Third, automated and standardized reporter assays are performed. We evaluated HiTmIR using miR-34a-5p, for which TNF- and TGFB-signaling, and Parkinson's Disease (PD)-related categories were identified and repeated the pipeline for miR-7-5p. HiTmIR validated 58.9% of the target genes for miR-34a-5p and 46.7% for miR-7-5p. We confirmed the targeting by measuring the endogenous protein levels of targets in a neuronal cell model. The standardized positive and negative targets are collected in the new miRATBase database, representing a resource for training, or benchmarking new target predictors. Applied to 88 target predictors with different confidence scores, TargetScan 7.2 and miRanda outperformed other tools. Our experiments demonstrate the efficiency of HiTmIR and provide evidence for an orchestrated miRNA-gene targeting.


Asunto(s)
Regulación de la Expresión Génica/genética , Ensayos Analíticos de Alto Rendimiento , MicroARNs/genética , 1-Metil-4-fenilpiridinio , Regiones no Traducidas 3' , Línea Celular , Línea Celular Tumoral , Genes Reporteros , Humanos , Mesencéfalo/citología , Neuroblastoma/patología , Neuronas/metabolismo , Enfermedad de Parkinson/genética , Valor Predictivo de las Pruebas , Sensibilidad y Especificidad , Transducción de Señal , Transcriptoma , Factor de Crecimiento Transformador beta/fisiología , Factor de Necrosis Tumoral alfa/fisiología
11.
Adv Exp Med Biol ; 1408: 309-328, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37093435

RESUMEN

Transforming growth factor-beta1 (TGF-ß) regulates a plethora of cell-intrinsic processes that modulate tumor progression in a context-dependent manner. Thus, although TGF-ß acts as a tumor suppressor in the early stages of tumorigenesis, in late stages, this factor promotes tumor progression and metastasis. In addition, TGF-ß also impinges on the tumor microenvironment by modulating the immune system. In this aspect, TGF-ß exhibits a potent immunosuppressive effect, which allows both cancer cells to escape from immune surveillance and confers resistance to immunotherapy. While TGF-ß inhibits the activation and antitumoral functions of T-cell lymphocytes, dendritic cells, and natural killer cells, it promotes the generation of T-regulatory cells and myeloid-derived suppressor cells, which hinder antitumoral T-cell activities. Moreover, TGF-ß promotes tumor-associated macrophages and neutrophils polarization from M1 into M2 and N1 to N2, respectively. Altogether, these effects contribute to the generation of an immunosuppressive tumor microenvironment and support tumor promotion. This review aims to analyze the relevant evidence on the complex role of TGF-ß in cancer immunology, the current outcomes of combined immunotherapies, and the anti-TGF-ß therapies that may improve the success of current and new oncotherapies.


Asunto(s)
Linfocitos T Reguladores , Factor de Crecimiento Transformador beta1 , Humanos , Células Asesinas Naturales , Carcinogénesis , Inmunoterapia , Factor de Crecimiento Transformador beta/fisiología , Microambiente Tumoral
12.
Gastroenterology ; 160(1): 245-259, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32941878

RESUMEN

BACKGROUND & AIMS: Mutations in the APC gene and other genes in the Wnt signaling pathway contribute to development of colorectal carcinomas. R-spondins (RSPOs) are secreted proteins that amplify Wnt signaling in intestinal stem cells. Alterations in RSPO genes have been identified in human colorectal tumors. We studied the effects of RSPO1 overexpression in ApcMin/+ mutant mice. METHODS: An adeno associated viral vector encoding RSPO1-Fc fusion protein, or control vector, was injected into ApcMin/+mice. Their intestinal crypts were isolated and cultured as organoids. which were incubated with or without RSPO1-Fc and an inhibitor of transforming growth factor beta receptor (TGFBR). Livers were collected from mice and analyzed by immunohistochemistry. Organoids and adenomas were analyzed by quantitative reverse-transcription PCR, single cell RNA sequencing, and immunohistochemistry. RESULTS: Intestines from Apc+/+ mice injected with the vector encoding RSPO1-Fc had significantly deeper crypts, longer villi, with increased EdU labeling, indicating increased proliferation of epithelial cells, in comparison to mice given control vector. AAV-RSPO1-Fc-transduced ApcMin/+ mice also developed fewer and smaller intestinal tumors and had significantly longer survival times. Adenomas of ApcMin/+ mice injected with the RSPO1-Fc vector showed a rapid increase in apoptosis and in the expression of Wnt target genes, followed by reduced expression of messenger RNAs and proteins regulated by the Wnt pathway, reduced cell proliferation, and less crypt branching than adenomas of mice given the control vector. Addition of RSPO1 reduced the number of adenoma organoids derived from ApcMin/+ mice and suppressed expression of Wnt target genes but increased phosphorylation of SMAD2 and transcription of genes regulated by SMAD. Inhibition of TGFBR signaling in organoids stimulated with RSPO1-Fc restored organoid formation and expression of genes regulated by Wnt. The TGFBR inhibitor restored apoptosis in adenomas from ApcMin/+ mice expressing RSPO1-Fc back to the same level as in the adenomas from mice given the control vector. CONCLUSIONS: Expression of RSPO1 in ApcMin/+ mice increases apoptosis and reduces proliferation and Wnt signaling in adenoma cells, resulting in development of fewer and smaller intestinal tumors and longer mouse survival. Addition of RSPO1 to organoids derived from adenomas inhibits their growth and promotes proliferation of intestinal stem cells that retain the APC protein; these effects are reversed by TGFB inhibitor. Strategies to increase the expression of RSPO1 might be developed for the treatment of intestinal adenomas.


Asunto(s)
Adenoma/patología , Neoplasias Intestinales/patología , Trombospondinas/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Vía de Señalización Wnt/fisiología , Adenoma/etiología , Animales , Modelos Animales de Enfermedad , Neoplasias Intestinales/etiología , Ratones , Organoides
13.
Mol Cell ; 56(6): 719-20, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25526529

RESUMEN

In this issue, Xu et al. (2014) show that innate antiviral RIG-I-like receptors (RLR) signaling represses TGF-ß-induced growth inhibition, epithelial-mesenchyme transition (EMT), and regulatory T cell (Treg) differentiation via IRF3-mediated Smads function.


Asunto(s)
Factor 3 Regulador del Interferón/fisiología , Virus Sendai/inmunología , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Humanos
14.
Mol Cell ; 56(6): 723-37, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25526531

RESUMEN

TGF-ß signaling is essential in many processes, including immune surveillance, and its dysregulation controls various diseases, including cancer, fibrosis, and inflammation. Studying the innate host defense, which functions in most cell types, we found that RLR signaling represses TGF-ß responses. This regulation is mediated by activated IRF3, using a dual mechanism of IRF3-directed suppression. Activated IRF3 interacts with Smad3, thus inhibiting TGF-ß-induced Smad3 activation and, in the nucleus, disrupts functional Smad3 transcription complexes by competing with coregulators. Consequently, IRF3 activation by innate antiviral signaling represses TGF-ß-induced growth inhibition, gene regulation and epithelial-mesenchymal transition, and the generation of Treg effector lymphocytes from naive CD4(+) lymphocytes. Conversely, silencing IRF3 expression enhances epithelial-mesenchymal transition, TGF-ß-induced Treg cell differentiation upon virus infection, and Treg cell generation in vivo. We present a mechanism of regulation of TGF-ß signaling by the antiviral defense, with evidence for its role in immune tolerance and cancer cell behavior.


Asunto(s)
Factor 3 Regulador del Interferón/fisiología , Virus Sendai/inmunología , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Diferenciación Celular , Transición Epitelial-Mesenquimal , Células HEK293 , Células Hep G2 , Humanos , Inmunidad Innata , Ratones Endogámicos C57BL , Transducción de Señal , Linfocitos T Reguladores/inmunología , Transcripción Genética , Activación Transcripcional/inmunología
15.
PLoS Genet ; 15(3): e1007706, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30849079

RESUMEN

In the natural environment, animals often encounter multiple sensory cues that are simultaneously present. The nervous system integrates the relevant sensory information to generate behavioral responses that have adaptive values. However, the neuronal basis and the modulators that regulate integrated behavioral response to multiple sensory cues are not well defined. Here, we address this question using a behavioral decision in C. elegans when the animal is presented with an attractive food source together with a repulsive odorant. We identify specific sensory neurons, interneurons and neuromodulators that orchestrate the decision-making process, suggesting that various states and contexts may modulate the multisensory integration. Among these modulators, we characterize a new function of a conserved TGF-ß pathway that regulates the integrated decision by inhibiting the signaling from a set of central neurons. Interestingly, we find that a common set of modulators, including the TGF-ß pathway, regulate the integrated response to the pairing of different foods and repellents. Together, our results provide mechanistic insights into the modulatory signals regulating multisensory integration.


Asunto(s)
Caenorhabditis elegans/fisiología , Animales , Animales Modificados Genéticamente , Conducta Animal/fisiología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/fisiología , Alimentos , Genes de Helminto , Interneuronas/fisiología , Cetonas , Mutación , Neuropéptidos/genética , Neuropéptidos/fisiología , Neurotransmisores/fisiología , Odorantes , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Células Receptoras Sensoriales/fisiología , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/fisiología
16.
Dev Dyn ; 250(7): 932-942, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33410237

RESUMEN

BACKGROUND: The transparent epidermis of Caenorhabditis elegans makes it an attractive model to study sperm motility and migration within an intact reproductive tract. C elegans synthesize specific F-series prostaglandins (PGFs) that are important for guiding sperm toward the spermatheca. These PGFs are synthesized from polyunsaturated fatty acid (PUFA) precursors, such as arachidonic acid (AA), via a novel pathway, independent of the classical cyclooxygenases (Cox) responsible for most PG synthesis. While the enzyme(s) responsible for PG synthesis has yet to be identified, the DAF-7 TGFß pathway has been implicated in modulating PG levels and sperm guidance. RESULTS: We find that the reduced PGF levels in daf-1 type I receptor mutants are responsible for the sperm guidance defect. The lower level of PGs in daf-1 mutants is due in part to the inaccessibility of AA. Finally, lipid analysis and assessment of sperm guidance in daf-1;daf-3 double mutants suggest DAF-3 suppresses PG production and sperm accumulation at the spermatheca. Our data suggest that DAF-3 functions in the nervous system, and possibly the germline, to affect sperm guidance. CONCLUSION: The C elegans TGFß pathway regulates many pathways to modulate PG metabolism and sperm guidance. These pathways likely function in the nervous system and possibly the germline.


Asunto(s)
Prostaglandinas/biosíntesis , Motilidad Espermática/genética , Factor de Crecimiento Transformador beta/fisiología , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Femenino , Masculino , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Motilidad Espermática/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
17.
Zhonghua Nan Ke Xue ; 28(9): 843-847, 2022 Sep.
Artículo en Zh | MEDLINE | ID: mdl-37839012

RESUMEN

Benign prostatic hyperplasia (BPH) is a most common disorder in the elderly. The development and growth of the prostate are regulated by androgens and growth factors, and the latter are secreted by prostatic epithelial and mesenchymal cells and play a key role. As the family members of FGF and TGF-ß, FGF2 and TGF-ß1 are the most representative growth factors in the prostate, involved in the regulation of such biological processes as proliferation, apoptosis, migration and differentiation of prostate cells, which provides a theoretical basis for the clinical application of FGF- and TGF-ß-targeted drugs in the treatment of BPH.


Asunto(s)
Hiperplasia Prostática , Factor de Crecimiento Transformador beta1 , Masculino , Humanos , Anciano , Factor de Crecimiento Transformador beta1/metabolismo , Hiperplasia Prostática/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Próstata/metabolismo , Factor de Crecimiento Transformador beta/fisiología
18.
J Biol Chem ; 295(27): 9033-9051, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32409577

RESUMEN

Cytochrome P450 1A1 (CYP1A1) catalyzes the metabolic activation of polycyclic aromatic hydrocarbons (PAHs) such as benzo[a]pyrene (B[a]P) and is transcriptionally regulated by the aryl hydrocarbon receptor (AhR)/AhR nuclear translocator (ARNT) complex upon exposure to PAHs. Accordingly, inhibition of CYP1A1 expression reduces production of carcinogens from PAHs. Although transcription of the CYP1A1 gene is known to be repressed by transforming growth factor-ß (TGF-ß), how TGF-ß signaling is involved in the suppression of CYP1A1 gene expression has yet to be clarified. In this study, using mammalian cell lines, along with shRNA-mediated gene silencing, CRISPR/Cas9-based genome editing, and reporter gene and quantitative RT-PCR assays, we found that TGF-ß signaling dissociates the B[a]P-mediated AhR/ARNT heteromeric complex. Among the examined Smads, Smad family member 3 (Smad3) strongly interacted with both AhR and ARNT via its MH2 domain. Moreover, hypoxia-inducible factor 1α (HIF-1α), which is stabilized upon TGF-ß stimulation, also inhibited AhR/ARNT complex formation in the presence of B[a]P. Thus, TGF-ß signaling negatively regulated the transcription of the CYP1A1 gene in at least two different ways. Of note, TGF-ß abrogated DNA damage in B[a]P-exposed cells. We therefore conclude that TGF-ß may protect cells against carcinogenesis because it inhibits CYP1A1-mediated metabolic activation of PAHs as part of its anti-tumorigenic activities.


Asunto(s)
Citocromo P-450 CYP1A1/genética , Receptores de Hidrocarburo de Aril/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células A549 , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Benzo(a)pireno/toxicidad , Células COS , Chlorocebus aethiops , Citocromo P-450 CYP1A1/metabolismo , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Hidrocarburos Policíclicos Aromáticos/metabolismo , Pirenos , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/fisiología
19.
Int J Cancer ; 148(5): 1245-1259, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33152121

RESUMEN

Tumour stromal cells support tumourigenesis. We report that Syndecan-2 (SDC2) is expressed on a nonepithelial, nonhaematopoietic, nonendothelial stromal cell population within breast cancer tissue. In vitro, syndecan-2 modulated TGFß signalling (SMAD7, PAI-1), migration and immunosuppression of patient-derived tumour-associated stromal cells (TASCs). In an orthotopic immunocompromised breast cancer model, overexpression of syndecan-2 in TASCs significantly enhanced TGFß signalling (SMAD7, PAI-1), tumour growth and metastasis, whereas reducing levels of SDC2 in TASCs attenuated TGFß signalling (SMAD7, PAI-1, CXCR4), tumour growth and metastasis. To explore the potential for therapeutic application, a syndecan-2-peptide was generated that inhibited the migratory and immunosuppressive properties of TASCs in association with reduced expression of TGFß-regulated immunosuppressive genes, such as CXCR4 and PD-L1. Moreover, using an orthotopic syngeneic breast cancer model, overexpression of syndecan-2-peptide in TASCs reduced tumour growth and immunosuppression within the TME. These data provide evidence that targeting stromal syndecan-2 within the TME inhibits tumour growth and metastasis due to decreased TGFß signalling and increased immune control.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Evasión Inmune , Sindecano-2/antagonistas & inhibidores , Animales , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Transducción de Señal/efectos de los fármacos , Células del Estroma/efectos de los fármacos , Células del Estroma/fisiología , Sindecano-2/fisiología , Factor de Crecimiento Transformador beta/fisiología , Microambiente Tumoral
20.
Cancer Sci ; 112(10): 4198-4207, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34375482

RESUMEN

Tumor-associated macrophages (TAMs), one of the most common cell components in the tumor microenvironment, have been reported as key contributors to cancer-related inflammation and enhanced metastatic progression of tumors. To explore the underlying mechanism of TAM-induced tumor progression, TAMs were isolated from colorectal cancer patients, and the functional interaction with colorectal cancer cells was analyzed. Our study found that coculture of TAMs contributed to a glycolytic state in colorectal cancer, which promoted the stem-like phenotypes and invasion of tumor cells. TAMs produced the cytokine transforming growth factor-ß to support hypoxia-inducible factor 1α (HIF1α) expression, thereby upregulating Tribbles pseudokinase 3 (TRIB3) in tumor cells. Elevated expression of TRIB3 resulted in activation of the ß-catenin/Wnt signaling pathway, which eventually enhanced the stem-like phenotypes and cell invasion in colorectal cancer. Our findings provided evidence that TAMs promoted colorectal cancer progression in a HIF1α/TRIB3-dependent manner, and blockade of HIF1α signals efficiently improved the outcome of chemotherapy, describing an innovative approach for colorectal cancer treatment.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Neoplasias Colorrectales/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Macrófagos Asociados a Tumores/fisiología , Animales , Proliferación Celular , Técnicas de Cocultivo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Femenino , Células HCT116 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica , Células Madre Neoplásicas , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral , Macrófagos Asociados a Tumores/metabolismo , Regulación hacia Arriba , Vía de Señalización Wnt/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA