Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Immunity ; 56(3): 606-619.e7, 2023 03 14.
Article in English | MEDLINE | ID: mdl-36750100

ABSTRACT

Although mice normally enter labor when their ovaries stop producing progesterone (luteolysis), parturition can also be triggered in this species through uterus-intrinsic pathways potentially analogous to the ones that trigger parturition in humans. Such pathways, however, remain largely undefined in both species. Here, we report that mice deficient in innate type 2 immunity experienced profound parturition delays when manipulated endocrinologically to circumvent luteolysis, thus obliging them to enter labor through uterus-intrinsic pathways. We found that these pathways were in part driven by the alarmin IL-33 produced by uterine interstitial fibroblasts. We also implicated important roles for uterine group 2 innate lymphoid cells, which demonstrated IL-33-dependent activation prior to labor onset, and eosinophils, which displayed evidence of elevated turnover in the prepartum uterus. These findings reveal a role for innate type 2 immunity in controlling the timing of labor onset through a cascade potentially relevant to human parturition.


Subject(s)
Interleukin-33 , Luteolysis , Pregnancy , Female , Mice , Animals , Humans , Interleukin-33/metabolism , Immunity, Innate , Myometrium/metabolism , Lymphocytes , Parturition/metabolism
2.
J Exp Med ; 219(5)2022 05 02.
Article in English | MEDLINE | ID: mdl-35416936

ABSTRACT

The paradox of fetomaternal tolerance has puzzled immunologists and reproductive biologists alike for almost 70 yr. Even the idea that the conceptus evokes a uniformly tolerogenic immune response in the mother is contradicted by the long-appreciated ability of pregnant women to mount robust antibody responses to paternal HLA molecules and RBC alloantigens such as Rh(D). Synthesizing these older observations with more recent work in mice, we discuss how the decision between tolerance or immunity to a given fetoplacental antigen appears to be a function of whether the antigen is trophoblast derived-and thus decorated with immunosuppressive glycans-or fetal blood cell derived.


Subject(s)
Fetal Blood , Trophoblasts , Animals , Female , Fetus , HLA Antigens , Humans , Immune Tolerance , Isoantigens , Mice , Pregnancy
3.
Cell Rep ; 38(5): 110329, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35108527

ABSTRACT

A little-appreciated feature of early pregnancy is that embryo implantation and placental outgrowth do not evoke wound-healing responses in the decidua, the specialized endometrial tissue that surrounds the conceptus. Here, we provide evidence that this phenomenon is partly due to an active program of gene silencing mediated by EZH2, a histone methyltransferase that generates repressive histone 3 lysine 27 trimethyl (H3K27me3) histone marks. We find that pregnancies in mice with EZH2-deficient decidual stromal cells frequently fail by mid-gestation, with the decidua showing ectopic myofibroblast formation, peri-embryonic collagen deposition, and gene expression profiles associated with transforming growth factor ß (TGF-ß)-driven fibroblast activation and fibrogenic extracellular matrix (ECM) remodeling. Analogous responses are observed when the mutant decidua is surgically wounded, while blockade of TGF-ß receptor signaling inhibits the defects and improves reproductive outcomes. Together, these results highlight a critical feature of reproductive success and have implications for the context-specific control of TGF-ß-mediated wound-healing responses elsewhere in the body.


Subject(s)
Embryo Implantation/physiology , Gene Silencing/physiology , Placenta/metabolism , Transforming Growth Factor beta/genetics , Wound Healing/physiology , Animals , Decidua/metabolism , Embryo, Mammalian/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Gene Expression/physiology , Histones/metabolism , Humans , Mice, Inbred C57BL , Pregnancy , Stromal Cells/metabolism , Transforming Growth Factor beta/metabolism
4.
J Clin Invest ; 130(1): 389-403, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31600172

ABSTRACT

Polymorphonuclear neutrophils (PMNs) are increasingly recognized to influence solid tumor development, but why their effects are so context dependent and even frequently divergent remains poorly understood. Using an autochthonous mouse model of uterine cancer and the administration of respiratory hyperoxia as a means to improve tumor oxygenation, we provide in vivo evidence that hypoxia is a potent determinant of tumor-associated PMN phenotypes and direct PMN-tumor cell interactions. Upon relief of tumor hypoxia, PMNs were recruited less intensely to the tumor-bearing uterus, but the recruited cells much more effectively killed tumor cells, an activity our data moreover suggested was mediated via their production of NADPH oxidase-derived reactive oxygen species and MMP-9. Simultaneously, their ability to promote tumor cell proliferation, which appeared to be mediated via their production of neutrophil elastase, was rendered less effective. Relieving tumor hypoxia thus greatly improved net PMN-dependent tumor control, leading to a massive reduction in tumor burden. Remarkably, this outcome was T cell independent. Together, these findings identify key hypoxia-regulated molecular mechanisms through which PMNs directly induce tumor cell death and proliferation in vivo and suggest that the contrasting properties of PMNs in different tumor settings may in part reflect the effects of hypoxia on direct PMN-tumor cell interactions.


Subject(s)
Cell Communication/immunology , Neoplasms, Experimental/immunology , Neutrophils/immunology , Uterine Neoplasms/immunology , Animals , Cell Hypoxia/immunology , Female , Mice , Mice, Knockout , Neoplasms, Experimental/pathology , Neutrophils/pathology , Uterine Neoplasms/pathology
5.
J Clin Invest ; 128(1): 233-247, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29202469

ABSTRACT

Uncovering the causes of pregnancy complications such as preterm labor requires greater insight into how the uterus remains in a noncontractile state until term and then surmounts this state to enter labor. Here, we show that dynamic generation and erasure of the repressive histone modification tri-methyl histone H3 lysine 27 (H3K27me3) in decidual stromal cells dictate both elements of pregnancy success in mice. In early gestation, H3K27me3-induced transcriptional silencing of select gene targets ensured uterine quiescence by preventing the decidua from expressing parturition-inducing hormone receptors, manifesting type 1 immunity, and most unexpectedly, generating myofibroblasts and associated wound-healing responses. In late gestation, genome-wide H3K27 demethylation allowed for target gene upregulation, decidual activation, and labor entry. Pharmacological inhibition of H3K27 demethylation in late gestation not only prevented term parturition, but also inhibited delivery while maintaining pup viability in a noninflammatory model of preterm parturition. Immunofluorescence analysis of human specimens suggested that similar regulatory events might occur in the human decidua. Together, these results reveal the centrality of regulated gene silencing in the uterine adaptation to pregnancy and suggest new areas in the study and treatment of pregnancy disorders.


Subject(s)
Decidua/metabolism , Histones/metabolism , Parturition/metabolism , Pregnancy/metabolism , Protein Processing, Post-Translational , Animals , Female , Gene Silencing/physiology , Humans , Male , Methylation , Mice , Up-Regulation/physiology
6.
Trends Mol Med ; 23(12): 1070-1071, 2017 12.
Article in English | MEDLINE | ID: mdl-29122491

ABSTRACT

Maternal immune activation (MIA) during pregnancy is associated with an increased risk of behavioral disorders in the offspring of affected mothers. Two recent studies highlight how maternal inflammation disrupts inhibitory interneuron networks and suggest that the maternal gut microbiome may be a contributing risk factor for MIA-induced behavioral abnormalities.


Subject(s)
Autistic Disorder/etiology , Brain/physiopathology , Gastrointestinal Microbiome/physiology , Inflammation/complications , Animals , Autistic Disorder/microbiology , Female , Inflammation/microbiology , Interneurons/physiology , Mothers , Pregnancy
7.
Infect Immun ; 85(8)2017 08.
Article in English | MEDLINE | ID: mdl-28507070

ABSTRACT

The bacterial pathogen Listeria monocytogenes causes foodborne systemic disease in pregnant women, which can lead to preterm labor, stillbirth, or severe neonatal disease. Colonization of the maternal decidua appears to be an initial step in the maternal component of the disease as well as bacterial transmission to the placenta and fetus. Host-pathogen interactions in the decidua during this early stage of infection remain poorly understood. Here, we assessed the dynamics of L. monocytogenes infection in primary human decidual organ cultures and in the murine decidua in vivo A high inoculum was necessary to infect both human and mouse deciduas, and the data support the existence of a barrier to initial colonization of the murine decidua. If successful, however, colonization in both species was followed by significant bacterial expansion associated with an inability of the decidua to mount appropriate innate cellular immune responses. The innate immune deficits included the failure of bacterial foci to attract macrophages and NK cells, cell types known to be important for early defenses against L. monocytogenes in the spleen, as well as a decrease in the tissue density of inflammatory Ly6Chi monocytes in vivo These results suggest that the infectivity of the decidua is not the result of an enhanced recruitment of L. monocytogenes to the gestational uterus but rather is due to compromised local innate cellular immune responses.


Subject(s)
Decidua/microbiology , Host-Pathogen Interactions , Listeria monocytogenes/immunology , Listeria monocytogenes/physiology , Listeriosis/immunology , Animals , Antigens, Ly/immunology , Decidua/immunology , Female , Humans , Immunity, Innate , Killer Cells, Natural/immunology , Listeria monocytogenes/growth & development , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Listeriosis/transmission , Macrophages/immunology , Mice , Monocytes/microbiology , Organ Culture Techniques , Placenta/immunology , Placenta/microbiology , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/microbiology , Spleen/immunology , Spleen/microbiology
8.
Sci Am ; 317(4): 46-53, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-29565852
9.
Cancer Cell ; 28(6): 785-799, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26678340

ABSTRACT

Polymorphonuclear neutrophils (PMNs) are largely considered to foster cancer development despite wielding an arsenal of cytotoxic agents. Using a mouse model of PTEN-deficient uterine cancer, we describe a surprising inhibitory role for PMNs in epithelial carcinogenesis. By inducing tumor cell detachment from the basement membrane, PMNs impeded early-stage tumor growth and retarded malignant progression. Unexpectedly, PMN recruitment and tumor growth control occurred independently of lymphocytes and cellular senescence and instead ensued as part of the tumor's intrinsic inflammatory response to hypoxia. In humans, a PMN gene signature correlated with improved survival in several cancer subtypes, including PTEN-deficient uterine cancer. These findings provide insight into tumor-associated PMNs and reveal a context-specific capacity for PMNs to directly combat tumorigenesis.


Subject(s)
Carcinoma, Endometrioid/prevention & control , Neutrophils/immunology , Ovarian Neoplasms/prevention & control , Phagocytosis , Tumor Microenvironment , Uterus/immunology , Animals , Bone Marrow Transplantation , Carcinoma, Endometrioid/enzymology , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/immunology , Carcinoma, Endometrioid/mortality , Carcinoma, Endometrioid/pathology , Cell Adhesion , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Chemotaxis , Computational Biology , Databases, Genetic , Female , Gene Expression Profiling , Gene Transfer Techniques , Humans , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Neoplasm Staging , Neutrophil Activation , Neutrophil Infiltration , Neutrophils/metabolism , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovariectomy , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Receptors, Colony-Stimulating Factor/deficiency , Receptors, Colony-Stimulating Factor/genetics , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Survival Analysis , Time Factors , Tumor Burden , Uterus/enzymology , Uterus/pathology
11.
Int J Dev Biol ; 58(2-4): 189-98, 2014.
Article in English | MEDLINE | ID: mdl-25023685

ABSTRACT

Understanding the function of T cells at the maternal-fetal interface remains one of the most difficult problems in reproductive immunology. A great deal of work over the last two decades has led to the view that the T cells that populate the decidua have important roles in both normal and pathological pregnancies, but the exact nature of these roles has remained unclear. Indeed, the old assumption that decidual T cells are uniformly threatening to fetal survival because the placenta is fundamentally an 'allograft' has given way to the idea that different T cell subsets contribute in different ways to pregnancy success or failure. Accordingly, some T cells are thought to protect the placenta from immune rejection and facilitate embryo implantation, while others are thought to contribute to pregnancy pathologies such as preeclampsia and spontaneous abortion. Here, we review the current state of information on the behavior of decidual T cells with a focus on both mouse and human studies, and with an emphasis on the many unresolved areas within this overall emerging framework.


Subject(s)
Decidua/immunology , Maternal-Fetal Exchange/immunology , Placenta/immunology , T-Lymphocytes/immunology , Animals , Female , Humans , Mice , Pregnancy
12.
Annu Rev Immunol ; 31: 387-411, 2013.
Article in English | MEDLINE | ID: mdl-23298207

ABSTRACT

The immune cells that reside at the interface between the placenta and uterus are thought to play many important roles in pregnancy. Recent work has revealed that the composition and function of these cells are locally controlled by the specialized uterine stroma (the decidua) that surrounds the implanted conceptus. Here, I discuss how key immune cell types (natural killer cells, macrophages, dendritic cells, and T cells) are either enriched or excluded from the decidua, how their function is regulated within the decidua, and how they variously contribute to pregnancy success or failure. The discussion emphasizes the relationship between human and mouse studies. Deeper understanding of the immunology of the maternal-fetal interface promises to yield significant insight into the pathogenesis of many human pregnancy complications, including preeclampsia, intrauterine growth restriction, spontaneous abortion, preterm birth, and congenital infection.


Subject(s)
Maternal-Fetal Exchange/immunology , Animals , Cell Differentiation/immunology , Decidua/cytology , Decidua/immunology , Decidua/pathology , Embryo Implantation/immunology , Female , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Placenta/blood supply , Placenta/immunology , Pregnancy
13.
Nat Rev Immunol ; 13(1): 23-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23237963

ABSTRACT

Work on the mechanisms of fetomaternal tolerance has undergone a renaissance in recent years, and the general outlines of a solution to this long-standing paradox of 'transplantation' immunology have come into view. Here, we discuss several mechanisms, recently described in mice, that either minimize the activation of maternal T cells with fetal or placental specificity, or minimize the possibility that such T cells, if activated, are able to harm the fetus. The T cell response to antigens expressed by the conceptus serves as a paradigm for the study of tissue-specific immune tolerance and is relevant to the pathogenesis of immune-mediated pregnancy complications.


Subject(s)
Fetus/immunology , Immune Tolerance , T-Lymphocytes/immunology , Animals , Female , Histocompatibility, Maternal-Fetal/immunology , Humans , Isoantigens , Lymphocyte Activation , Male , Maternal-Fetal Exchange/immunology , Mice , Models, Immunological , Pregnancy , T-Lymphocytes, Regulatory/immunology , Uterus/anatomy & histology , Uterus/immunology
14.
PLoS One ; 8(12): e84064, 2013.
Article in English | MEDLINE | ID: mdl-24391885

ABSTRACT

Maternal immune tolerance towards the fetus and placenta is thought to be established in part by pathways that attenuate T cell priming to antigens released from the placenta into maternal blood. These pathways remain largely undefined and their existence, at face value, seems incompatible with a mother's need to maintain a functional immune system during pregnancy. A particular conundrum is evident if we consider that maternal antigen presenting cells, activated in order to prime T cells to pathogen-derived antigens, would also have the capacity to prime T cells to co-ingested placental antigens. Here, we address this paradox using a transgenic system in which placental membranes are tagged with a strong surrogate antigen (ovalbumin). We find that although a remarkably large quantity of acellular ovalbumin-containing placental material is released into maternal blood, splenic CD8 T cells in pregnant mice bearing unmanipulated T cell repertoires are not primed to ovalbumin even if the mice are intravenously injected with adjuvants. This failure was largely independent of regulatory T cells, and instead was linked to the intrinsic characteristics of the released material that rendered it selectively non-immunogenic, potentially by sequestering it from CD8α(+) dendritic cells. The release of ovalbumin-containing placental material into maternal blood thus had no discernable impact on CD8 T cell priming to soluble ovalbumin injected intravenously during pregnancy, nor did it induce long-term tolerance to ovalbumin. Together, these results outline a major pathway governing the maternal immune response to the placenta, and suggest how tolerance to placental antigens can be maintained systemically without being detrimental to host defense.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Fetus/immunology , Immune Tolerance/immunology , Ovalbumin/immunology , Placenta/immunology , Receptors, CCR7/physiology , T-Lymphocytes, Regulatory/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Dendritic Cells/immunology , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy
15.
Science ; 336(6086): 1317-21, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22679098

ABSTRACT

The chemokine-mediated recruitment of effector T cells to sites of inflammation is a central feature of the immune response. The extent to which chemokine expression levels are limited by the intrinsic developmental characteristics of a tissue has remained unexplored. We show in mice that effector T cells cannot accumulate within the decidua, the specialized stromal tissue encapsulating the fetus and placenta. Impaired accumulation was in part attributable to the epigenetic silencing of key T cell-attracting inflammatory chemokine genes in decidual stromal cells, as evidenced by promoter accrual of repressive histone marks. These findings give insight into mechanisms of fetomaternal immune tolerance, as well as reveal the epigenetic modification of tissue stromal cells as a modality for limiting effector T cell trafficking.


Subject(s)
Chemokines/genetics , Decidua/immunology , Decidua/metabolism , Gene Silencing , Immune Tolerance , Stromal Cells/immunology , Stromal Cells/metabolism , T-Lymphocyte Subsets/immunology , Animals , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL11/genetics , Chemokine CXCL11/metabolism , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Chemokines/metabolism , Chromatin Immunoprecipitation , Endometrium/cytology , Endometrium/immunology , Female , Histones/metabolism , Immunologic Memory , Inflammation , Methylation , Mice , Mice, Inbred C57BL , Myometrium/immunology , Ovalbumin/immunology , Pregnancy , Promoter Regions, Genetic , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism
17.
Ann N Y Acad Sci ; 1276: 37-46, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23278645

ABSTRACT

Sponsored by the New York Academy of Sciences and Cincinnati Children's Hospital Medical Center, with support from the National Institute of Environmental Health Sciences (NIEHS), the National Institute on Drug Abuse (NIDA), and Life Technologies, "Fetal Programming and Environmental Exposures: Implications for Prenatal Care and Preterm Birth" was held on June 11-12, 2012 at the New York Academy of Sciences in New York City. The meeting, comprising individual talks and panel discussions, highlighted basic, clinical, and translational research approaches, and highlighted the need for specialized testing of drugs, consumer products, and industrial chemicals, with a view to the unique impacts these can have during gestation. Speakers went on to discuss many other factors that affect prenatal development, from genetics to parental diet, revealing the extraordinary sensitivity of the developing fetus.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Environmental Exposure/adverse effects , Fetal Development , Endocrine Disruptors/adverse effects , Female , Fetal Development/genetics , Humans , Pregnancy , Premature Birth , Prenatal Care , Translational Research, Biomedical
18.
Expert Rev Clin Immunol ; 7(5): 593-602, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21895472

ABSTRACT

Understanding the evolutionary adaptation of the immune system to the developing fetus and placenta represents one of the most fascinating problems in reproductive biology. Recent work has focused on how the behavior of dendritic cells (DCs) is altered at the maternal-fetal interface to suit the unique requirements of pregnancy. This work has provided a significant new perspective into the long-standing immunological paradox of fetomaternal tolerance, and has opened up a new and intriguing area of research into the potential trophic role of uterine DCs in the peri-implantation period. Further research on the biology of uterine DCs promises to give insight into the pathogenesis of many clinically important disorders of pregnancy.


Subject(s)
Decidua/immunology , Dendritic Cells/immunology , Immune Tolerance , Maternal-Fetal Exchange/immunology , Animals , Decidua/pathology , Dendritic Cells/pathology , Female , Humans , Male , Pregnancy , Pregnancy Complications/immunology , Pregnancy Complications/pathology
19.
J Exp Med ; 208(9): 1901-16, 2011 Aug 29.
Article in English | MEDLINE | ID: mdl-21825019

ABSTRACT

Tissue macrophages (Mφs) and dendritic cells (DCs) play essential roles in tissue homeostasis and immunity. How these cells are maintained at their characteristic densities in different tissues has remained unclear. Aided by a novel flow cytometric technique for assessing relative rates of blood-borne precursor recruitment, we examined Mφ and DC population dynamics in the pregnant mouse uterus, where rapid tissue growth facilitated a dissection of underlying regulatory mechanisms. We demonstrate how Mφ dynamics, and thus Mφ tissue densities, are locally controlled by CSF-1, a pleiotropic growth factor whose in situ level of activity varied widely between uterine tissue layers. CSF-1 acted in part by inducing Mφ proliferation and in part by stimulating the extravasation of Ly6C(hi) monocytes (Mos) that served as Mφ precursors. Mo recruitment was dependent on the production of CCR2 chemokine receptor ligands by uterine Mφs in response to CSF-1. Unexpectedly, a parallel CSF-1-regulated, but CCR2-independent pathway influenced uterine DC tissue densities by controlling local pre-DC extravasation rates. Together, these data provide cellular and molecular insight into the regulation of Mφ tissue densities under noninflammatory conditions and reveal a central role for CSF-1 in the coordination of Mφ and DC homeostasis.


Subject(s)
Cell Movement/immunology , Cell Proliferation , Dendritic Cells/immunology , Macrophage Colony-Stimulating Factor/immunology , Macrophages/immunology , Pregnancy/immunology , Uterus/immunology , Animals , Dendritic Cells/cytology , Female , Macrophage Colony-Stimulating Factor/genetics , Macrophages/cytology , Mice , Mice, Knockout , Monocytes/immunology , Pregnancy/genetics , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Uterus/cytology
20.
Cancer Cell ; 20(1): 104-18, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21741600

ABSTRACT

To metastasize, a tumor cell must acquire abilities such as the capacity to colonize new tissue and evade immune surveillance. Recent evidence suggests that microRNAs can promote the evolution of malignant behaviors by regulating multiple targets. We performed a microRNA analysis of human melanoma, a highly invasive cancer, and found that miR-30b/30d upregulation correlates with stage, metastatic potential, shorter time to recurrence, and reduced overall survival. Ectopic expression of miR-30b/30d promoted the metastatic behavior of melanoma cells by directly targeting the GalNAc transferase GALNT7, resulted in increased synthesis of the immunosuppressive cytokine IL-10, and reduced immune cell activation and recruitment. These data support a key role of miR-30b/30d and GalNAc transferases in metastasis, by simultaneously promoting cellular invasion and immunosuppression.


Subject(s)
Immune Tolerance/genetics , MicroRNAs/metabolism , N-Acetylgalactosaminyltransferases/metabolism , Animals , Base Sequence , Cell Proliferation , Disease Progression , Gene Expression Regulation, Neoplastic , Gene Silencing , Genome, Human/genetics , Humans , Interleukin-10/metabolism , Melanoma/enzymology , Melanoma/genetics , Melanoma/pathology , Mice , MicroRNAs/genetics , Molecular Sequence Data , N-Acetylgalactosaminyltransferases/antagonists & inhibitors , N-Acetylgalactosaminyltransferases/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Polysaccharides/metabolism , Up-Regulation , Polypeptide N-acetylgalactosaminyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL
...