Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Science ; 378(6621): eabl7207, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36395212

ABSTRACT

Many human cancers manifest the capability to circumvent attack by the adaptive immune system. In this work, we identified a component of immune evasion that involves frequent up-regulation of fragile X mental retardation protein (FMRP) in solid tumors. FMRP represses immune attack, as revealed by cancer cells engineered to lack its expression. FMRP-deficient tumors were infiltrated by activated T cells that impaired tumor growth and enhanced survival in mice. Mechanistically, FMRP's immunosuppression was multifactorial, involving repression of the chemoattractant C-C motif chemokine ligand 7 (CCL7) concomitant with up-regulation of three immunomodulators-interleukin-33 (IL-33), tumor-secreted protein S (PROS1), and extracellular vesicles. Gene signatures associate FMRP's cancer network with poor prognosis and response to therapy in cancer patients. Collectively, FMRP is implicated as a regulator that orchestrates a multifaceted barrier to antitumor immune responses.


Subject(s)
Fragile X Mental Retardation Protein , Immune Evasion , Immune Tolerance , Neoplasms , Animals , Humans , Mice , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Neoplasms/immunology , Chemokine CCL7/metabolism , Interleukin-33 , Protein S/metabolism
2.
Nat Commun ; 11(1): 1571, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32218432

ABSTRACT

Estrogens and progesterone control breast development and carcinogenesis via their cognate receptors expressed in a subset of luminal cells in the mammary epithelium. How they control the extracellular matrix, important to breast physiology and tumorigenesis, remains unclear. Here we report that both hormones induce the secreted protease Adamts18 in myoepithelial cells by controlling Wnt4 expression with consequent paracrine canonical Wnt signaling activation. Adamts18 is required for stem cell activation, has multiple binding partners in the basement membrane and interacts genetically with the basal membrane-specific proteoglycan, Col18a1, pointing to the basement membrane as part of the stem cell niche. In vitro, ADAMTS18 cleaves fibronectin; in vivo, Adamts18 deletion causes increased collagen deposition during puberty, which results in impaired Hippo signaling and reduced Fgfr2 expression both of which control stem cell function. Thus, Adamts18 links luminal hormone receptor signaling to basement membrane remodeling and stem cell activation.


Subject(s)
ADAMTS Proteins/metabolism , Hormones/pharmacology , Mammary Glands, Animal/cytology , Stem Cell Niche , Stem Cells/metabolism , ADAMTS Proteins/deficiency , ADAMTS Proteins/genetics , Animals , Antigens, CD/metabolism , Cell Line , Cell Self Renewal/drug effects , Epithelium/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Fibronectins/metabolism , Glycoproteins/metabolism , Humans , Mice, Inbred C57BL , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Progesterone/metabolism , Regeneration/drug effects , Signal Transduction/drug effects , Stem Cell Niche/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Transcription, Genetic/drug effects
3.
Nat Commun ; 9(1): 4723, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30413705

ABSTRACT

Oestrogen receptor α (ERα) is a transcription factor with ligand-independent and ligand-dependent activation functions (AF)-1 and -2. Oestrogens control postnatal mammary gland development acting on a subset of mammary epithelial cells (MECs), termed sensor cells, which are ERα-positive by immunohistochemistry (IHC) and secrete paracrine factors, which stimulate ERα-negative responder cells. Here we show that deletion of AF-1 or AF-2 blocks pubertal ductal growth and subsequent development because both are required for expression of essential paracrine mediators. Thirty percent of the luminal cells are ERα-negative by IHC but express Esr1 transcripts. This low level ERα expression through AF-2 is essential for cell expansion during puberty and growth-inhibitory during pregnancy. Cell-intrinsic ERα is not required for cell proliferation nor for secretory differentiation but controls transcript levels of cell motility and cell adhesion genes and a stem cell and epithelial mesenchymal transition (EMT) signature identifying ERα as a key regulator of mammary epithelial cell plasticity.


Subject(s)
Epithelium/metabolism , Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Mammary Glands, Animal/metabolism , Animals , Cell Proliferation , Endocrine System/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation , Mammary Glands, Animal/growth & development , Mice, Inbred C57BL , Phenotype , Pregnancy , Protein Domains , RNA, Messenger/genetics , RNA, Messenger/metabolism , Steroids/metabolism , Structure-Activity Relationship
4.
Biol Open ; 5(11): 1585-1594, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27638769

ABSTRACT

The ADAMTS family comprises 19 secreted metalloproteinases that cleave extracellular matrix components and have diverse functions in numerous disease and physiological contexts. A number of them remain 'orphan' proteases and among them is ADAMTS18, which has been implicated in developmental eye disorders, platelet function and various malignancies. To assess in vivo function of ADAMTS18, we generated a mouse strain with inactivated Adamts18 alleles. In the C57Bl6/Ola background, Adamts18-deficient mice are born in a normal Mendelian ratio, and are viable but show a transient growth delay. Histological examination revealed a 100% penetrant eye defect resulting from leakage of lens material through the lens capsule occurring at embryonic day (E)13.5, when the lens grows rapidly. Adamts18-deficient lungs showed altered bronchiolar branching. Fifty percent of mutant females are infertile because of vaginal obstruction due to either a dorsoventral vaginal septum or imperforate vagina. The incidence of ovarian rete is increased in the mutant mouse strain. Thus, Adamts18 is essential in the development of distinct tissues and the new mouse strain is likely to be useful for investigating ADAMTS18 function in human disease, particularly in the contexts of infertility and carcinogenesis.

5.
Wiley Interdiscip Rev Dev Biol ; 4(3): 181-95, 2015.
Article in English | MEDLINE | ID: mdl-25645332

ABSTRACT

Most of mammary gland development occurs postnatally under the control of female reproductive hormones, which in turn interact with other endocrine factors. While hormones impinge on many tissues and trigger very complex biological responses, tissue recombination experiments with hormone receptor-deficient mammary epithelia revealed eminent roles for estrogens, progesterone, and prolactin receptor (PrlR) signaling that are intrinsic to the mammary epithelium. A subset of the luminal mammary epithelial cells expresses the estrogen receptor α (ERα), the progesterone receptor (PR), and the PrlR and act as sensor cells. These cells convert the detected systemic signals into local signals that are developmental stage-dependent and may be direct, juxtacrine, or paracrine. This setup ensures that the original input is amplified and that the biological responses of multiple cell types can be coordinated. Some key mediators of hormone action have been identified such as Wnt, EGFR, IGFR, and RANK signaling. Multiple signaling pathways such as FGF, Hedgehog, and Notch signaling participate in driving different aspects of mammary gland development locally but how they link to the hormonal control remains to be elucidated. An increasing number of endocrine factors are appearing to have a role in mammary gland development, the adipose tissue is increasingly recognized to play a role in endocrine regulation, and a complex role of the immune system with multiple different cell types is being revealed. For further resources related to this article, please visit the WIREs website.


Subject(s)
Endocrine System/physiology , Epithelial Cells/metabolism , Mammary Glands, Animal/growth & development , Models, Biological , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Receptors, Prolactin/metabolism , Signal Transduction/physiology , Animals , Female , Mammary Glands, Animal/metabolism , Mice
6.
J Biol Chem ; 289(39): 26960-26972, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25107905

ABSTRACT

Somatic mutations altering lysine 171 of the IKBKB gene that encodes (IKKß), the critical activating kinase in canonical (NFκB) signaling, have been described in splenic marginal zone lymphomas and multiple myeloma. Lysine 171 forms part of a cationic pocket that interacts with the activation loop phosphate in the activated wild type kinase. We show here that K171E IKKß and K171T IKKß represent kinases that are constitutively active even in the absence of activation loop phosphorylation. Predictive modeling and biochemical studies establish why mutations in a positively charged residue in the cationic pocket of an activation loop phosphorylation-dependent kinase result in constitutive activation. Transcription activator-like effector nuclease-based knock-in mutagenesis provides evidence from a B lymphoid context that K171E IKKß contributes to lymphomagenesis.


Subject(s)
I-kappa B Kinase , Lymphoma , Mutation, Missense , NF-kappa B , Neoplasm Proteins , Signal Transduction/genetics , Amino Acid Substitution , HeLa Cells , Humans , I-kappa B Kinase/immunology , I-kappa B Kinase/metabolism , Lymphoma/genetics , Lymphoma/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphorylation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...