Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 147
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(9): 2454-2470.e26, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33857425

RESUMEN

Glioblastoma multiforme (GBM) is an aggressive brain tumor for which current immunotherapy approaches have been unsuccessful. Here, we explore the mechanisms underlying immune evasion in GBM. By serially transplanting GBM stem cells (GSCs) into immunocompetent hosts, we uncover an acquired capability of GSCs to escape immune clearance by establishing an enhanced immunosuppressive tumor microenvironment. Mechanistically, this is not elicited via genetic selection of tumor subclones, but through an epigenetic immunoediting process wherein stable transcriptional and epigenetic changes in GSCs are enforced following immune attack. These changes launch a myeloid-affiliated transcriptional program, which leads to increased recruitment of tumor-associated macrophages. Furthermore, we identify similar epigenetic and transcriptional signatures in human mesenchymal subtype GSCs. We conclude that epigenetic immunoediting may drive an acquired immune evasion program in the most aggressive mesenchymal GBM subtype by reshaping the tumor immune microenvironment.


Asunto(s)
Neoplasias Encefálicas/inmunología , Epigénesis Genética , Glioblastoma/inmunología , Evasión Inmune/inmunología , Células Mieloides/inmunología , Células Madre Neoplásicas/inmunología , Microambiente Tumoral/inmunología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular , Metilación de ADN , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Mieloides/metabolismo , Células Mieloides/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Immunity ; 54(5): 885-902, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33979586

RESUMEN

Tumor cells metastasize to distant organs through a complex series of events that are driven by tumor intrinsic and extrinsic factors. In particular, non-malignant stromal cells, including immune cells, modify tumor metastatic behavior. Of these cells, tumor-associated innate immune cells, particularly macrophages and neutrophils, suppress the cytotoxic activity of innate and adaptive killer cells and interact with tumor cells to promote their growth and malignancy. These findings in mouse cancer models suggest that targeting these sub-populations of immune cells holds therapeutic promise in treating metastatic disease. In this review, we describe the origin and role of the macrophages, neutrophils, and their progenitors in the metastatic cascade and suggest strategies that might enhance cancer therapy.


Asunto(s)
Macrófagos/inmunología , Metástasis de la Neoplasia/inmunología , Neutrófilos/inmunología , Animales , Biología/métodos , Humanos , Células Asesinas Naturales/inmunología , Neoplasias/inmunología , Células del Estroma/inmunología
3.
Immunity ; 47(2): 217-218, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28813653

RESUMEN

In this issue of Immunity, Zhu et al. (2017) report that tumor-associated macrophages in a mouse model of pancreatic ductal adenocarcinoma (PDAC) originate from both the yolk sac (YS) and bone marrow. Differential ablation of these populations indicates that only the YS-derived macrophages promote PDAC progression and growth.


Asunto(s)
Carcinoma Ductal Pancreático , Saco Vitelino , Animales , Médula Ósea , Macrófagos , Páncreas
4.
Cell ; 141(1): 39-51, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20371344

RESUMEN

There is persuasive clinical and experimental evidence that macrophages promote cancer initiation and malignant progression. During tumor initiation, they create an inflammatory environment that is mutagenic and promotes growth. As tumors progress to malignancy, macrophages stimulate angiogenesis, enhance tumor cell migration and invasion, and suppress antitumor immunity. At metastatic sites, macrophages prepare the target tissue for arrival of tumor cells, and then a different subpopulation of macrophages promotes tumor cell extravasation, survival, and subsequent growth. Specialized subpopulations of macrophages may represent important new therapeutic targets.


Asunto(s)
Macrófagos/patología , Metástasis de la Neoplasia , Neoplasias/inmunología , Neoplasias/patología , Animales , Progresión de la Enfermedad , Humanos , Macrófagos/inmunología , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología
5.
Trends Immunol ; 42(2): 104-119, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33446416

RESUMEN

Monocytes are progenitors to macrophages and a subclass of dendritic cells (monocyte-derived dendritic cells, MoDCs), but they also act as circulating sensors that respond to environmental changes and disease. Technological advances have defined the production of classical monocytes in the bone marrow through the identification of lineage-determining transcription factors (LDTFs) and have proposed alternative routes of differentiation. Monocytes released into the circulation can be recruited to tissues by specific chemoattractants where they respond to sequential niche-specific signals that determine their differentiation into terminal effector cells. New aspects of monocyte biology in the circulation are being revealed, exemplified by the influence of cancer on the systemic alteration of monocyte subset abundance and transcriptional profiles. These changes can act to enhance the metastatic spread of primary cancers and may offer therapeutic opportunities.


Asunto(s)
Monocitos , Neoplasias , Diferenciación Celular , Células Dendríticas , Homeostasis , Humanos , Macrófagos
6.
Nature ; 562(7726): 223-228, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30258231

RESUMEN

The earliest blood vessels in mammalian embryos are formed when endothelial cells differentiate from angioblasts and coalesce into tubular networks. Thereafter, the endothelium is thought to expand solely by proliferation of pre-existing endothelial cells. Here we show that a complementary source of endothelial cells is recruited into pre-existing vasculature after differentiation from the earliest precursors of erythrocytes, megakaryocytes and macrophages, the erythro-myeloid progenitors (EMPs) that are born in the yolk sac. A first wave of EMPs contributes endothelial cells to the yolk sac endothelium, and a second wave of EMPs colonizes the embryo and contributes endothelial cells to intraembryonic endothelium in multiple organs, where they persist into adulthood. By demonstrating that EMPs constitute a hitherto unrecognized source of endothelial cells, we reveal that embryonic blood vascular endothelium expands in a dual mechanism that involves both the proliferation of pre-existing endothelial cells and the incorporation of endothelial cells derived from haematopoietic precursors.


Asunto(s)
Vasos Sanguíneos/citología , Vasos Sanguíneos/embriología , Linaje de la Célula , Células Endoteliales/citología , Eritrocitos/citología , Células Progenitoras Mieloides/citología , Envejecimiento , Animales , Linaje de la Célula/genética , Proliferación Celular , Células Endoteliales/metabolismo , Eritrocitos/metabolismo , Perfilación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Integrasas/genética , Integrasas/metabolismo , Hígado/citología , Hígado/embriología , Ratones , Células Progenitoras Mieloides/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Rombencéfalo/irrigación sanguínea , Rombencéfalo/citología , Rombencéfalo/embriología , Transcripción Genética , Saco Vitelino/citología , Saco Vitelino/embriología
7.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33536334

RESUMEN

Macrophages are intimately involved in the pathophysiology of endometriosis, a chronic inflammatory disorder characterized by the growth of endometrial-like tissue (lesions) outside the uterus. By combining genetic and pharmacological monocyte and macrophage depletion strategies we determined the ontogeny and function of macrophages in a mouse model of induced endometriosis. We demonstrate that lesion-resident macrophages are derived from eutopic endometrial tissue, infiltrating large peritoneal macrophages (LpM) and monocytes. Furthermore, we found endometriosis to trigger continuous recruitment of monocytes and expansion of CCR2+ LpM. Depletion of eutopic endometrial macrophages results in smaller endometriosis lesions, whereas constitutive inhibition of monocyte recruitment significantly reduces peritoneal macrophage populations and increases the number of lesions. Reprogramming the ontogeny of peritoneal macrophages such that embryo-derived LpM are replaced by monocyte-derived LpM decreases the number of lesions that develop. We propose a putative model whereby endometrial macrophages are "proendometriosis" while newly recruited monocyte-derived macrophages, possibly in LpM form, are "antiendometriosis." These observations highlight the importance of monocyte-derived macrophages in limiting disease progression.


Asunto(s)
Endometriosis/patología , Macrófagos Peritoneales/patología , Animales , Anticuerpos Monoclonales/metabolismo , Quimiocina CCL2/deficiencia , Quimiocina CCL2/metabolismo , Endometrio/patología , Femenino , Ratones Endogámicos C57BL , Modelos Biológicos , Monocitos/patología , Cavidad Peritoneal/patología
8.
Immunity ; 41(1): 49-61, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25035953

RESUMEN

The tumor microenvironment is a complex ecology of cells that evolves with and provides support to tumor cells during the transition to malignancy. Among the innate and adaptive immune cells recruited to the tumor site, macrophages are particularly abundant and are present at all stages of tumor progression. Clinical studies and experimental mouse models indicate that these macrophages generally play a protumoral role. In the primary tumor, macrophages can stimulate angiogenesis and enhance tumor cell invasion, motility, and intravasation. During monocytes and/or metastasis, macrophages prime the premetastatic site and promote tumor cell extravasation, survival, and persistent growth. Macrophages are also immunosuppressive, preventing tumor cell attack by natural killer and T cells during tumor progression and after recovery from chemo- or immunotherapy. Therapeutic success in targeting these protumoral roles in preclinical models and in early clinical trials suggests that macrophages are attractive targets as part of combination therapy in cancer treatment.


Asunto(s)
Linaje de la Célula/inmunología , Transformación Celular Neoplásica/inmunología , Macrófagos/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Humanos , Terapia de Inmunosupresión , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/patología , Neovascularización Patológica/inmunología , Microambiente Tumoral
9.
Liver Int ; 43(1): 208-220, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35924447

RESUMEN

BACKGROUND AND AIMS: Dickkopf-1 (DKK1) is associated with poor prognosis in intrahepatic cholangiocarcinoma (iCCA), but the mechanisms behind this are unclear. Here, we show that DKK1 plays an immune regulatory role in vivo and inhibition reduces tumour growth. METHODS: Various in vivo GEMM mouse models and patient samples were utilized to assess the effects of tumour specific DKK1 overexpression in iCCA. DKK1-driven changes to the tumour immune microenvironment were characterized by immunostaining and gene expression analysis. DKK1 overexpressing and damage-induced models of iCCA were used to demonstrate the therapeutic efficacy of DKK1 inhibition in these contexts using the anti-DKK1 therapeutic, DKN-01. RESULTS: DKK1 overexpression in mouse models of iCCA drives an increase in chemokine and cytokine signalling, the recruitment of regulatory macrophages, and promotes the formation of a tolerogenic niche with higher numbers of regulatory T cells. We show a similar association of DKK1 with FOXP3 and regulatory T cells in patient tissue and gene expression data, demonstrating these effects are relevant to human iCCA. Finally, we demonstrate that inhibition of DKK1 with the monoclonal antibody mDKN-01 is effective at reducing tumour burden in two distinct mouse models of the disease. CONCLUSION: DKK1 promotes tumour immune evasion in iCCA through the recruitment of immune suppressive macrophages. Targeting DKK1 with a neutralizing antibody is effective at reducing tumour growth in vivo. As such, DKK1 targeted and immune modulatory therapies may be an effective strategy in iCCA patients with high DKK1 tumour expression or tolerogenic immune phenotypes.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Péptidos y Proteínas de Señalización Intercelular , Animales , Humanos , Ratones , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/metabolismo , Conductos Biliares Intrahepáticos/patología , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Fenotipo , Microambiente Tumoral
10.
Proc Natl Acad Sci U S A ; 116(9): 3604-3613, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30733286

RESUMEN

Cancer cells have higher reactive oxygen species (ROS) than normal cells, due to genetic and metabolic alterations. An emerging scenario is that cancer cells increase ROS to activate protumorigenic signaling while activating antioxidant pathways to maintain redox homeostasis. Here we show that, in basal-like and BRCA1-related breast cancer (BC), ROS levels correlate with the expression and activity of the transcription factor aryl hydrocarbon receptor (AhR). Mechanistically, ROS triggers AhR nuclear accumulation and activation to promote the transcription of both antioxidant enzymes and the epidermal growth factor receptor (EGFR) ligand, amphiregulin (AREG). In a mouse model of BRCA1-related BC, cancer-associated AhR and AREG control tumor growth and production of chemokines to attract monocytes and activate proangiogenic function of macrophages in the tumor microenvironment. Interestingly, the expression of these chemokines as well as infiltration of monocyte-lineage cells (monocyte and macrophages) positively correlated with ROS levels in basal-like BC. These data support the existence of a coordinated link between cancer-intrinsic ROS regulation and the features of tumor microenvironment. Therapeutically, chemical inhibition of AhR activity sensitizes human BC models to Erlotinib, a selective EGFR tyrosine kinase inhibitor, suggesting a promising combinatorial anticancer effect of AhR and EGFR pathway inhibition. Thus, AhR represents an attractive target to inhibit redox homeostasis and modulate the tumor promoting microenvironment of basal-like and BRCA1-associated BC.


Asunto(s)
Anfirregulina/genética , Proteína BRCA1/genética , Neoplasias de la Mama/genética , Receptores de Hidrocarburo de Aril/genética , Adulto , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Receptores ErbB/genética , Clorhidrato de Erlotinib/administración & dosificación , Femenino , Regulación Neoplásica de la Expresión Génica , Homeostasis/genética , Humanos , Ratones , Persona de Mediana Edad , Oxidación-Reducción/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Microambiente Tumoral/genética
11.
Immunity ; 36(6): 1031-46, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22749353

RESUMEN

GM-CSF (Csf-2) is a critical cytokine for the in vitro generation of dendritic cells (DCs) and is thought to control the development of inflammatory DCs and resident CD103(+) DCs in some tissues. Here we showed that in contrast to the current understanding, Csf-2 receptor acts in the steady state to promote the survival and homeostasis of nonlymphoid tissue-resident CD103(+) and CD11b(+) DCs. Absence of Csf-2 receptor on lung DCs abrogated the induction of CD8(+) T cell immunity after immunization with particulate antigens. In contrast, Csf-2 receptor was dispensable for the differentiation and innate function of inflammatory DCs during acute injuries. Instead, inflammatory DCs required Csf-1 receptor for their development. Thus, Csf-2 is important in vaccine-induced CD8(+) T cell immunity through the regulation of nonlymphoid tissue DC homeostasis rather than control of inflammatory DCs in vivo.


Asunto(s)
Subunidad beta Común de los Receptores de Citocinas/fisiología , Células Dendríticas/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Inflamación/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Linaje de la Célula , Subunidad beta Común de los Receptores de Citocinas/antagonistas & inhibidores , Subunidad beta Común de los Receptores de Citocinas/deficiencia , Subunidad beta Común de los Receptores de Citocinas/genética , Células Dendríticas/clasificación , Células Dendríticas/citología , Encefalomielitis Autoinmune Experimental/inmunología , Endotoxemia/inmunología , Perfilación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Homeostasis , Lipopolisacáridos/toxicidad , Listeriosis/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/trasplante , Especificidad de Órganos , Infecciones por Orthomyxoviridae/inmunología , Infecciones Neumocócicas/inmunología , Quimera por Radiación , Bazo/inmunología , Tamoxifeno/farmacología
12.
Cancer Immunol Immunother ; 68(4): 687-697, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30684003

RESUMEN

In cancer, infection and inflammation, the immune system's function can be dysregulated. Instead of fighting disease, immune cells may increase pathology and suppress host-protective immune responses. Myeloid cells show high plasticity and adapt to changing conditions and pathological challenges. Despite their relevance in disease pathophysiology, the identity, heterogeneity and biology of myeloid cells is still poorly understood. We will focus on phenotypical and functional markers of one of the key myeloid regulatory subtypes, the myeloid derived suppressor cells (MDSC), in humans, mice and non-human primates. Technical issues regarding the isolation of the cells from tissues and blood, timing and sample handling of MDSC will be detailed. Localization of MDSC in a tissue context is of crucial importance and immunohistochemistry approaches for this purpose are discussed. A minimal antibody panel for MDSC research is provided as part of the Mye-EUNITER COST action. Strategies for the identification of additional markers applying state of the art technologies such as mass cytometry will be highlighted. Such marker sets can be used to study MDSC phenotypes across tissues, diseases as well as species and will be crucial to accelerate MDSC research in health and disease.


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Animales , Biomarcadores , Separación Celular/métodos , Humanos , Inmunofenotipificación/métodos , Ratones , Neutrófilos/inmunología , Neutrófilos/metabolismo , Primates
13.
Nature ; 496(7446): 445-55, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23619691

RESUMEN

Macrophages, the most plastic cells of the haematopoietic system, are found in all tissues and show great functional diversity. They have roles in development, homeostasis, tissue repair and immunity. Although tissue macrophages are anatomically distinct from one another, and have different transcriptional profiles and functional capabilities, they are all required for the maintenance of homeostasis. However, these reparative and homeostatic functions can be subverted by chronic insults, resulting in a causal association of macrophages with disease states. In this Review, we discuss how macrophages regulate normal physiology and development, and provide several examples of their pathophysiological roles in disease. We define the 'hallmarks' of macrophages according to the states that they adopt during the performance of their various roles, taking into account new insights into the diversity of their lineages, identities and regulation. It is essential to understand this diversity because macrophages have emerged as important therapeutic targets in many human diseases.


Asunto(s)
Enfermedad , Crecimiento y Desarrollo , Homeostasis , Macrófagos/patología , Macrófagos/fisiología , Animales , Linaje de la Célula , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Macrófagos/citología , Macrófagos/inmunología
14.
Angew Chem Int Ed Engl ; 58(47): 16894-16898, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31535788

RESUMEN

We report the novel chemical design of fluorescent activatable chemokines as highly specific functional probes for imaging subpopulations of immune cells in live tumours. Activatable chemokines behave as AND-gates since they emit only after receptor binding and intracellular activation, showing enhanced selectivity over existing agents. We have applied this strategy to produce mCCL2-MAF as the first probe for in vivo detection of metastasis-associated macrophages in a preclinical model of lung metastasis. This strategy will accelerate the preparation of new chemokine-based probes for imaging immune cell function in tumours.


Asunto(s)
Neoplasias de la Mama/patología , Colorantes Fluorescentes/química , Neoplasias Pulmonares/patología , Macrófagos/patología , Imagen Molecular/métodos , Receptores CCR2/fisiología , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Proliferación Celular , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Espectrometría de Fluorescencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Proc Natl Acad Sci U S A ; 112(11): E1382-91, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25733860

RESUMEN

The uterine epithelium of mice and humans undergoes cyclical waves of cell proliferation and differentiation under the regulation of estradiol-17ß (E2) and progesterone (P4). These epithelial cells respond to E2 with increased protein and DNA synthesis, whereas P4 inhibits only the E2-induced DNA synthetic response. Here we show that E2 regulates protein synthesis in these epithelial cells through activating PKC that in turn stimulates ERK1/2 to phosphorylate and thereby activate the central regulator of protein synthesis mechanistic target of rapamycin (mTOR). This mTOR pathway is not inhibited by P4. Inhibitor studies with an estrogen receptor (ESR1) antagonist showed the dependence of this mTOR pathway on ESR1 but that once activated, a phosphorylation cascade independent of ESR1 propagates the pathway. E2 also stimulates an IGF1 receptor (IGF1R) to PI3 kinase to AKT to GSK-3ß pathway required for activation of the canonical cell cycle machinery that is inhibited by P4. PKC activation did not stimulate this pathway nor does inhibition of PKC or ERK1/2 affect it. These studies therefore indicate a mechanism whereby DNA and protein synthesis are regulated by two ESR1-activated pathways that run in parallel with only the one responsible for the initiation of DNA synthesis blocked by P4. Inhibition of mTOR by rapamycin in vivo resulted in inhibition of E2-induced protein and DNA synthesis. Proliferative diseases of the endometrium such as endometriosis and cancer are common and E2 dependent. Thus, defining this mTOR pathway suggests that local (intrauterine or peritoneal) rapamycin administration might be a therapeutic option for these diseases.


Asunto(s)
Células Epiteliales/metabolismo , Estradiol/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Proteína Quinasa C/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Útero/citología , Animales , Activación Enzimática/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Ratones , Modelos Biológicos , Progesterona/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
16.
Differentiation ; 98: 62-69, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29156254

RESUMEN

The human endometrium undergoes extensive morphological, biochemical and molecular changes under the influence of female sex steroid hormones. Besides the fact that estrogen stimulates endometrial cell proliferation and progesterone inhibits this proliferation and induces differentiation, there is limited knowledge about precise molecular mechanisms underlying human endometrial biology. The importance of paracrine signaling in endometrial physiology explains why in vitro culture of endometrial cells has been challenging. Researchers, therefore, have developed alternative experimental in vivo models for the study of endometrial biology. The objective of this review is to summarize the recent developments and work on these in vivo endometrial research models. The in vivo recombinant tissue models in which wild-type endometrial cells are combined with endometrial cells from a gene-targeted mouse strain followed by xenografting to host mice have been critical in confirming the significance of paracrine signaling between the epithelium and stroma in the growth regulation of the endometrium. Additionally, these studies have uncovered differences between the mouse and human, emphasizing the need for the development of experimental models specifically of the human endometrium. Recently, xenotransplants of human endometrial fragments into the subcutaneous space of host mice and endometrial xenografts of dissociated and recombined epithelial and stromal cells beneath the kidney capsule of immunodeficient host mice have proven to be highly promising tools for in vivo research of endometrial functions. For the first time, the latter approach provides an immense opportunity for the application of genome engineering, such as targeted ablation of endometrial genes for example by using CRISPR/CAS9 system. This research will begin to elucidate the functional role of specific genes in this complex tissue. Another advantage of xenotransplantation and xenograft models of the human endometrium is their use to investigate endometrial effects of new compounds and drugs without needing to give them to women. Underpinning the molecular mechanisms underlying endometrial functions is critical to ultimately advance our understanding of endometrial pathophysiology and develop targeted therapies to prevent and cure endometrial pathologies as well as enhance endometrial function when it is desired for fertility.


Asunto(s)
Diferenciación Celular/fisiología , Endometrio/citología , Células Epiteliales/citología , Células del Estroma/citología , Animales , Proliferación Celular/fisiología , Epitelio/patología , Femenino , Humanos
17.
Breast Cancer Res ; 19(1): 4, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28077158

RESUMEN

BACKGROUND: Macrophages play diverse roles in mammary gland development and breast cancer. CC-chemokine ligand 2 (CCL2) is an inflammatory cytokine that recruits macrophages to sites of injury. Although CCL2 has been detected in human and mouse mammary epithelium, its role in regulating mammary gland development and cancer risk has not been explored. METHODS: Transgenic mice were generated wherein CCL2 is driven by the mammary epithelial cell-specific mouse mammary tumour virus 206 (MMTV) promoter. Estrous cycles were tracked in adult transgenic and non-transgenic FVB mice, and mammary glands collected at the four different stages of the cycle. Dissected mammary glands were assessed for cyclical morphological changes, proliferation and apoptosis of epithelium, macrophage abundance and collagen deposition, and mRNA encoding matrix remodelling enzymes. Another cohort of control and transgenic mice received carcinogen 7,12-Dimethylbenz(a)anthracene (DMBA) and tumour development was monitored weekly. CCL2 protein was also quantified in paired samples of human breast tissue with high and low mammographic density. RESULTS: Overexpression of CCL2 in the mammary epithelium resulted in an increased number of macrophages, increased density of stroma and collagen and elevated mRNA encoding matrix remodelling enzymes lysyl oxidase (LOX) and tissue inhibitor of matrix metalloproteinases (TIMP)3 compared to non-transgenic controls. Transgenic mice also exhibited increased susceptibility to development of DMBA-induced mammary tumours. In a paired sample cohort of human breast tissue, abundance of epithelial-cell-associated CCL2 was higher in breast tissue of high mammographic density compared to tissue of low mammographic density. CONCLUSIONS: Constitutive expression of CCL2 by the mouse mammary epithelium induces a state of low level chronic inflammation that increases stromal density and elevates cancer risk. We propose that CCL2-driven inflammation contributes to the increased risk of breast cancer observed in women with high mammographic density.


Asunto(s)
Neoplasias de la Mama/etiología , Neoplasias de la Mama/metabolismo , Quimiocina CCL2/metabolismo , Susceptibilidad a Enfermedades , Inflamación/metabolismo , Células del Estroma/metabolismo , Animales , Densidad de la Mama , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Quimiocina CCL2/genética , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Epitelio/metabolismo , Ciclo Estral , Femenino , Expresión Génica , Humanos , Inflamación/genética , Inflamación/patología , Estimación de Kaplan-Meier , Macrófagos/inmunología , Macrófagos/metabolismo , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Neoplasias Mamarias Experimentales , Ratones , Ratones Transgénicos , Pronóstico , ARN Mensajero/genética , Células del Estroma/patología
18.
Nature ; 475(7355): 222-5, 2011 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-21654748

RESUMEN

Macrophages, which are abundant in the tumour microenvironment, enhance malignancy. At metastatic sites, a distinct population of metastasis-associated macrophages promotes the extravasation, seeding and persistent growth of tumour cells. Here we define the origin of these macrophages by showing that Gr1-positive inflammatory monocytes are preferentially recruited to pulmonary metastases but not to primary mammary tumours in mice. This process also occurs for human inflammatory monocytes in pulmonary metastases of human breast cancer cells. The recruitment of these inflammatory monocytes, which express CCR2 (the receptor for chemokine CCL2), as well as the subsequent recruitment of metastasis-associated macrophages and their interaction with metastasizing tumour cells, is dependent on CCL2 synthesized by both the tumour and the stroma. Inhibition of CCL2-CCR2 signalling blocks the recruitment of inflammatory monocytes, inhibits metastasis in vivo and prolongs the survival of tumour-bearing mice. Depletion of tumour-cell-derived CCL2 also inhibits metastatic seeding. Inflammatory monocytes promote the extravasation of tumour cells in a process that requires monocyte-derived vascular endothelial growth factor. CCL2 expression and macrophage infiltration are correlated with poor prognosis and metastatic disease in human breast cancer. Our data provide the mechanistic link between these two clinical associations and indicate new therapeutic targets for treating metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Quimiocina CCL2/metabolismo , Inflamación/patología , Monocitos/patología , Metástasis de la Neoplasia , Animales , Antígeno CD11b/metabolismo , Quimiocina CCL2/antagonistas & inhibidores , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Receptores de Lipopolisacáridos/metabolismo , Neoplasias Pulmonares/secundario , Macrófagos/patología , Ratones , Monocitos/metabolismo , Metástasis de la Neoplasia/tratamiento farmacológico , Trasplante de Neoplasias , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Receptores CCR2/antagonistas & inhibidores , Receptores CCR2/metabolismo , Receptores de IgG/metabolismo , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Nature ; 474(7352): 511-5, 2011 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-21623369

RESUMEN

Myeloid cells are a feature of most tissues. Here we show that during development, retinal myeloid cells (RMCs) produce Wnt ligands to regulate blood vessel branching. In the mouse retina, where angiogenesis occurs postnatally, somatic deletion in RMCs of the Wnt ligand transporter Wntless results in increased angiogenesis in the deeper layers. We also show that mutation of Wnt5a and Wnt11 results in increased angiogenesis and that these ligands elicit RMC responses via a non-canonical Wnt pathway. Using cultured myeloid-like cells and RMC somatic deletion of Flt1, we show that an effector of Wnt-dependent suppression of angiogenesis by RMCs is Flt1, a naturally occurring inhibitor of vascular endothelial growth factor (VEGF). These findings indicate that resident myeloid cells can use a non-canonical, Wnt-Flt1 pathway to suppress angiogenic branching.


Asunto(s)
Células Mieloides/metabolismo , Neovascularización Fisiológica/fisiología , Retina/citología , Transducción de Señal , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Wnt/metabolismo , Animales , Vasos Sanguíneos/crecimiento & desarrollo , Células Endoteliales/metabolismo , Fibroblastos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas Relacionadas con Receptor de LDL/metabolismo , Ligandos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Receptores Acoplados a Proteínas G , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 1 de Factores de Crecimiento Endotelial Vascular/deficiencia , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Proteínas Wnt/deficiencia , Proteínas Wnt/genética , Proteína Wnt-5a
20.
Gastroenterology ; 149(7): 1896-1909.e14, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26344055

RESUMEN

BACKGROUND & AIMS: Liver regeneration requires functional liver macrophages, which provide an immune barrier that is compromised after liver injury. The numbers of liver macrophages are controlled by macrophage colony-stimulating factor (CSF1). We examined the prognostic significance of the serum level of CSF1 in patients with acute liver injury and studied its effects in mice. METHODS: We measured levels of CSF1 in serum samples collected from 55 patients who underwent partial hepatectomy at the Royal Infirmary Edinburgh between December 2012 and October 2013, as well as from 78 patients with acetaminophen-induced acute liver failure admitted to the Royal Infirmary Edinburgh or the University of Kansas Medical Centre. We studied the effects of increased levels of CSF1 in uninjured mice that express wild-type CSF1 receptor or a constitutive or inducible CSF1-receptor reporter, as well as in chemokine receptor 2 (Ccr2)-/- mice; we performed fate-tracing experiments using bone marrow chimeras. We administered CSF1-Fc (fragment, crystallizable) to mice after partial hepatectomy and acetaminophen intoxication, and measured regenerative parameters and innate immunity by clearance of fluorescent microbeads and bacterial particles. RESULTS: Serum levels of CSF1 increased in patients undergoing liver surgery in proportion to the extent of liver resected. In patients with acetaminophen-induced acute liver failure, a low serum level of CSF1 was associated with increased mortality. In mice, administration of CSF1-Fc promoted hepatic macrophage accumulation via proliferation of resident macrophages and recruitment of monocytes. CSF1-Fc also promoted transdifferentiation of infiltrating monocytes into cells with a hepatic macrophage phenotype. CSF1-Fc increased innate immunity in mice after partial hepatectomy or acetaminophen-induced injury, with resident hepatic macrophage as the main effector cells. CONCLUSIONS: Serum CSF1 appears to be a prognostic marker for patients with acute liver injury. CSF1 might be developed as a therapeutic agent to restore innate immune function after liver injury.


Asunto(s)
Transdiferenciación Celular , Factores Estimulantes de Colonias , Animales , Humanos , Inmunidad Innata , Hígado/efectos de los fármacos , Fallo Hepático Agudo/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA