Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Autophagy ; : 1-23, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38963021

ABSTRACT

The commonality between various muscle diseases is the loss of muscle mass, function, and regeneration, which severely restricts mobility and impairs the quality of life. With muscle stem cells (MuSCs) playing a key role in facilitating muscle repair, targeting regulators of muscle regeneration has been shown to be a promising therapeutic approach to repair muscles. However, the underlying molecular mechanisms driving muscle regeneration are complex and poorly understood. Here, we identified a new regulator of muscle regeneration, Deaf1 (Deformed epidermal autoregulatory factor-1) - a transcriptional factor downstream of foxo signaling. We showed that Deaf1 is transcriptionally repressed by FOXOs and that DEAF1 targets to Pik3c3 and Atg16l1 promoter regions and suppresses their expression. Deaf1 depletion therefore induces macroautophagy/autophagy, which in turn blocks MuSC survival and differentiation. In contrast, Deaf1 overexpression inactivates autophagy in MuSCs, leading to increased protein aggregation and cell death. The fact that Deaf1 depletion and its overexpression both lead to defects in muscle regeneration highlights the importance of fine tuning DEAF1-regulated autophagy during muscle regeneration. We further showed that Deaf1 expression is altered in aging and cachectic MuSCs. Manipulation of Deaf1 expression can attenuate muscle atrophy and restore muscle regeneration in aged mice or mice with cachectic cancers. Together, our findings unveil an evolutionarily conserved role for DEAF1 in muscle regeneration, providing insights into the development of new therapeutic strategies against muscle atrophy.Abbreviations: DEAF1: Deformed epidermal autoregulatory factor-1; FOXO: Forkhead box O; MuSC: Muscle Stem Cell; PAX7: Paired box 7; PIK3C3: Phosphatidylinositol 3-kinase catalytic subunit type 3.

2.
Nat Commun ; 15(1): 36, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167296

ABSTRACT

While canonical Wnt signaling is well recognized for its crucial regulatory functions in cell fate decisions, the role of non-canonical Wnt signaling in adult stem cells remains elusive and contradictory. Here, we identified Mcam, a potential member of the non-canonical Wnt signaling, as an important negative regulator of mammary gland epithelial cells (MECs) by genome-scale CRISPR-Cas9 knockout (GeCKO) library screening. Loss of Mcam increases the clonogenicity and regenerative capacity of MECs, and promotes the proliferation, differentiation, and ductal morphogenesis of mammary epithelial in knockout mice. Mechanically, Mcam knockout recruits and polarizes macrophages through the Il4-Stat6 axis, thereby promoting secretion of the non-canonical Wnt ligand Wnt5a and its binding to the non-canonical Wnt signaling receptor Ryk to induce the above phenotypes. These findings reveal Mcam roles in mammary gland development by orchestrating communications between MECs and macrophages via a Wnt5a/Ryk axis, providing evidences for non-canonical Wnt signaling in mammary development.


Subject(s)
Wnt Proteins , Wnt Signaling Pathway , Mice , Animals , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism , Cell Differentiation , Morphogenesis , Mice, Knockout , Macrophages/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
3.
Cells ; 13(2)2024 01 19.
Article in English | MEDLINE | ID: mdl-38275818

ABSTRACT

Tetraspanins, a superfamily of small integral membrane proteins, are characterized by four transmembrane domains and conserved protein motifs that are configured into a unique molecular topology and structure in the plasma membrane. They act as key organizers of the plasma membrane, orchestrating the formation of specialized microdomains called "tetraspanin-enriched microdomains (TEMs)" or "tetraspanin nanodomains" that are essential for mediating diverse biological processes. TSPAN8 is one of the earliest identified tetraspanin members. It is known to interact with a wide range of molecular partners in different cellular contexts and regulate diverse molecular and cellular events at the plasma membrane, including cell adhesion, migration, invasion, signal transduction, and exosome biogenesis. The functions of cell-surface TSPAN8 are governed by ER targeting, modifications at the Golgi apparatus and dynamic trafficking. Intriguingly, limited evidence shows that TSPAN8 can translocate to the nucleus to act as a transcriptional regulator. The transcription of TSPAN8 is tightly regulated and restricted to defined cell lineages, where it can serve as a molecular marker of stem/progenitor cells in certain normal tissues as well as tumors. Importantly, the oncogenic roles of TSPAN8 in tumor development and cancer metastasis have gained prominence in recent decades. Here, we comprehensively review the current knowledge on the molecular characteristics and regulatory mechanisms defining TSPAN8 functions, and discuss the potential and significance of TSPAN8 as a biomarker and therapeutic target across various epithelial cancers.


Subject(s)
Neoplasms , Tetraspanins , Humans , Tetraspanins/genetics , Tetraspanins/metabolism , Neoplasms/genetics , Membrane Proteins , Cell Membrane/metabolism , Cell Adhesion
4.
Cell ; 186(10): 2144-2159.e22, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37172565

ABSTRACT

Bats are special in their ability to live long and host many emerging viruses. Our previous studies showed that bats have altered inflammasomes, which are central players in aging and infection. However, the role of inflammasome signaling in combating inflammatory diseases remains poorly understood. Here, we report bat ASC2 as a potent negative regulator of inflammasomes. Bat ASC2 is highly expressed at both the mRNA and protein levels and is highly potent in inhibiting human and mouse inflammasomes. Transgenic expression of bat ASC2 in mice reduced the severity of peritonitis induced by gout crystals and ASC particles. Bat ASC2 also dampened inflammation induced by multiple viruses and reduced mortality of influenza A virus infection. Importantly, it also suppressed SARS-CoV-2-immune-complex-induced inflammasome activation. Four key residues were identified for the gain of function of bat ASC2. Our results demonstrate that bat ASC2 is an important negative regulator of inflammasomes with therapeutic potential in inflammatory diseases.


Subject(s)
Apoptosis Regulatory Proteins , Chiroptera , Inflammasomes , Ribonucleoproteins , Virus Diseases , Animals , Humans , Mice , Apoptosis Regulatory Proteins/metabolism , Chiroptera/immunology , COVID-19 , Inflammasomes/immunology , Ribonucleoproteins/metabolism , SARS-CoV-2 , Virus Diseases/immunology , Virus Physiological Phenomena
5.
Cell Rep ; 42(2): 112065, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36724073

ABSTRACT

Tetraspanins, a superfamily of membrane proteins, mediate diverse biological processes through tetraspanin-enriched microdomains in the plasma membrane. However, how their cell-surface presentation is controlled remains unclear. To identify the regulators of tetraspanin trafficking, we conduct sequential genome-wide loss-of-function CRISPR-Cas9 screens based on cell-surface expression of a tetraspanin member, TSPAN8. Several genes potentially involved in endoplasmic reticulum (ER) targeting, different biological processes in the Golgi apparatus, and protein trafficking are identified and functionally validated. Importantly, we find that biantennary N-glycans generated by MGAT1/2, but not more complex glycan structures, are important for cell-surface tetraspanin expression. Moreover, we unravel that SPPL3, a Golgi intramembrane-cleaving protease reported previously to act as a sheddase of multiple glycan-modifying enzymes, controls cell-surface tetraspanin expression through a mechanism associated with lacto-series glycolipid biosynthesis. Our study provides critical insights into the molecular regulation of cell-surface presentation of tetraspanins with implications for strategies to manipulate their functions, including cancer cell invasion.


Subject(s)
CRISPR-Cas Systems , Neoplasms , Humans , CRISPR-Cas Systems/genetics , Tetraspanins/genetics , Tetraspanins/metabolism , Cell Membrane/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasms/genetics
6.
Front Cell Dev Biol ; 9: 704547, 2021.
Article in English | MEDLINE | ID: mdl-34336857

ABSTRACT

The ability to regulate the survival and death of a cell is paramount throughout the lifespan of a multicellular organism. Apoptosis, a main physiological form of programmed cell death, is regulated by the Bcl-2 family proteins that are either pro-apoptotic or pro-survival. The in vivo functions of distinct Bcl-2 family members are largely unmasked by genetically engineered murine models. Mcl-1 is one of the two Bcl-2 like pro-survival genes whose germline deletion causes embryonic lethality in mice. Its requisite for the survival of a broad range of cell types has been further unraveled by using conditional and inducible deletion murine model systems in different tissues or cell lineages and at distinct developmental stages. Moreover, genetic mouse cancer models have also demonstrated that Mcl-1 is essential for the survival of multiple tumor types. The MCL-1 locus is commonly amplified across various cancer types in humans. Small molecule inhibitors with high affinity and specificity to human MCL-1 have been developed and explored for the treatment of certain cancers. To facilitate the pre-clinical studies of MCL-1 in cancer and other diseases, transgenic mouse models over-expressing human MCL-1 as well as humanized MCL-1 mouse models have been recently engineered. This review discusses the current advances in understanding the physiological roles of Mcl-1 based on studies using genetic murine models and its critical implications in pathology and treatment of human diseases.

7.
EMBO Rep ; 22(1): e50854, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33393215

ABSTRACT

Nrf2 signaling is vital for protecting cells against oxidative stress. However, its hyperactivation is frequently found in liver cancer through excessive build-up of p62/SQSTM1 bodies that sequester Keap1, an adaptor of the E3-ubiquitin ligase complex for Nrf2. Here, we report that the Bax-binding protein MOAP-1 regulates p62-Keap1-Nrf2 signaling through disruption of p62 bodies. Upon induction of cellular stresses that stimulate formation of p62 bodies, MOAP-1 is recruited to p62 bodies and reduces their levels independent of the autophagy pathway. MOAP-1 interacts with the PB1-ZZ domains of p62 and interferes with its self-oligomerization and liquid-liquid phase separation, thereby disassembling the p62 bodies. Loss of MOAP-1 can lead to marked upregulation of p62 bodies, enhanced sequestration of Keap1 by p62 and hyperactivation of Nrf2 antioxidant target genes. MOAP-1-deficient mice exhibit an elevated tumor burden with excessive levels of p62 bodies and Nrf2 signaling in a diethylnitrosamine (DEN)-induced hepatocarcinogenesis model. Together, our data define MOAP-1 as a negative regulator of Nrf2 signaling via dissociation of p62 bodies.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2 , Signal Transduction , Animals , Autophagy , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism
8.
Physiol Rev ; 100(2): 489-523, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31539305

ABSTRACT

The mammary gland is a highly dynamic organ that undergoes profound changes within its epithelium during puberty and the reproductive cycle. These changes are fueled by dedicated stem and progenitor cells. Both short- and long-lived lineage-restricted progenitors have been identified in adult tissue as well as a small pool of multipotent mammary stem cells (MaSCs), reflecting intrinsic complexity within the epithelial hierarchy. While unipotent progenitor cells predominantly execute day-to-day homeostasis and postnatal morphogenesis during puberty and pregnancy, multipotent MaSCs have been implicated in coordinating alveologenesis and long-term ductal maintenance. Nonetheless, the multipotency of stem cells in the adult remains controversial. The advent of large-scale single-cell molecular profiling has revealed striking changes in the gene expression landscape through ontogeny and the presence of transient intermediate populations. An increasing number of lineage cell-fate determination factors and potential niche regulators have now been mapped along the hierarchy, with many implicated in breast carcinogenesis. The emerging diversity among stem and progenitor populations of the mammary epithelium is likely to underpin the heterogeneity that characterizes breast cancer.


Subject(s)
Cell Differentiation , Cell Lineage , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Developmental , Humans , Mammary Glands, Animal/pathology , Mammary Glands, Human/pathology , Morphogenesis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Signal Transduction , Stem Cells/pathology , Transcription Factors/genetics , Tumor Microenvironment
9.
Proc Natl Acad Sci U S A ; 116(39): 19530-19540, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31488716

ABSTRACT

Emerging evidence suggests that hepatocytes are primarily maintained by self-renewal during normal liver homeostasis, as well as in response to a wide variety of hepatic injuries. However, how hepatocytes in distinct anatomic locations within the liver lobule are replenished under homeostasis and injury-induced regeneration remains elusive. Using a newly developed bacterial artificial chromosome (BAC)-transgenic mouse model, we demonstrate that Lgr5 expression in the liver is restricted to a unique subset of hepatocytes most adjacent to the central veins. Genetic lineage tracing revealed that pericentral Lgr5+ hepatocytes have a long lifespan and mainly contribute to their own lineage maintenance during postnatal liver development and homeostasis. Remarkably, these hepatocytes also fuel the regeneration of their own lineage during the massive and rapid regeneration process following two-thirds partial hepatectomy. Moreover, Lgr5+ hepatocytes are found to be the main cellular origin of diethylnitrosamine (DEN)-induced hepatocellular carcinoma (HCC) and are highly susceptible to neoplastic transformation triggered by activation of Erbb pathway. Our findings establish an unexpected self-maintaining mode for a defined subset of hepatocytes during liver homeostasis and regeneration, and identify Lgr5+ pericentral hepatocytes as major cells of origin in HCC development.


Subject(s)
Hepatocytes/physiology , Liver Regeneration/physiology , Receptors, G-Protein-Coupled/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Proliferation/physiology , Disease Models, Animal , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Liver/physiology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Transgenic , Stem Cells/cytology
11.
Cancer Cell ; 35(4): 618-632.e6, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30930118

ABSTRACT

Breast tumors are inherently heterogeneous, but the evolving cellular organization through neoplastic progression is poorly understood. Here we report a rapid, large-scale single-cell resolution 3D imaging protocol based on a one-step clearing agent that allows visualization of normal tissue architecture and entire tumors at cellular resolution. Imaging of multicolor lineage-tracing models of breast cancer targeted to either basal or luminal progenitor cells revealed profound clonal restriction during progression. Expression profiling of clones arising in Pten/Trp53-deficient tumors identified distinct molecular signatures. Strikingly, most clones harbored cells that had undergone an epithelial-to-mesenchymal transition, indicating widespread, inherent plasticity. Hence, an integrative pipeline that combines lineage tracing, 3D imaging, and clonal RNA sequencing technologies offers a comprehensive path for studying mechanisms underlying heterogeneity in whole tumors.


Subject(s)
Breast Neoplasms/pathology , Cell Lineage , Cell Plasticity , Epithelial-Mesenchymal Transition , Imaging, Three-Dimensional , Microscopy, Confocal , Single-Cell Analysis/methods , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Lineage/genetics , Cell Plasticity/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Humans , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Sequence Analysis, RNA , Transcriptome , Tumor Burden
12.
Dev Cell ; 47(5): 629-644.e8, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30523786

ABSTRACT

Long-lived quiescent mammary stem cells (MaSCs) are presumed to coordinate the dramatic expansion of ductal epithelium that occurs through the different phases of postnatal development, but little is known about the molecular regulators that underpin their activation. We show that ablation of the transcription factor Foxp1 in the mammary gland profoundly impairs ductal morphogenesis, resulting in a rudimentary tree throughout life. Foxp1-deficient glands were highly enriched for quiescent Tspan8hi MaSCs, which failed to become activated even in competitive transplantation assays, thus highlighting a cell-intrinsic defect. Foxp1 deletion also resulted in aberrant expression of basal genes in luminal cells, inferring a role in cell-fate decisions. Notably, Foxp1 was uncovered as a direct repressor of Tspan8 in basal cells, and deletion of Tspan8 rescued the defects in ductal morphogenesis elicited by Foxp1 loss. Thus, a single transcriptional regulator Foxp1 can control the exit of MaSCs from dormancy to orchestrate differentiation and development.


Subject(s)
Adult Stem Cells/metabolism , Cell Differentiation , Forkhead Transcription Factors/metabolism , Mammary Glands, Human/growth & development , Morphogenesis , Repressor Proteins/metabolism , 3T3 Cells , Adult Stem Cells/cytology , Animals , Cells, Cultured , Female , Forkhead Transcription Factors/genetics , HEK293 Cells , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Mice , Mice, Inbred C57BL , Repressor Proteins/genetics , Tetraspanins/genetics , Tetraspanins/metabolism
13.
Blood ; 130(23): 2504-2515, 2017 12 07.
Article in English | MEDLINE | ID: mdl-28972012

ABSTRACT

T-cell differentiation is governed by interactions with thymic epithelial cells (TECs) and defects in this process undermine immune function and tolerance. To uncover new strategies to restore thymic function and adaptive immunity in immunodeficiency, we sought to determine the molecular mechanisms that control life and death decisions in TECs. Guided by gene expression profiling, we created mouse models that specifically deleted prosurvival genes in TECs. We found that although BCL-2 and BCL-XL were dispensable for TEC homeostasis, MCL-1 deficiency impacted on TECs as early as embryonic day 15.5, resulting in early thymic atrophy and T-cell lymphopenia, with near complete loss of thymic tissue by 2 months of age. MCL-1 was not necessary for TEC differentiation but was continually required for the survival of mature cortical and medullary TECs and the maintenance of thymic architecture. A screen of TEC trophic factors in organ cultures showed that epidermal growth factor upregulated MCL-1 via MAPK/ERK kinase activity, providing a molecular mechanism for the support of TEC survival. This signaling axis governing TEC survival and thymic function represents a new target for strategies for thymic protection and regeneration.


Subject(s)
Cell Survival/genetics , Epithelial Cells/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Thymus Gland/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/drug effects , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Female , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Homeostasis/genetics , Immunophenotyping , Lymphopenia/genetics , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism , Thymus Gland/pathology , bcl-X Protein/genetics , bcl-X Protein/metabolism
14.
Nat Cell Biol ; 19(3): 164-176, 2017 03.
Article in English | MEDLINE | ID: mdl-28192422

ABSTRACT

Despite accumulating evidence for a mammary differentiation hierarchy, the basal compartment comprising stem cells remains poorly characterized. Through gene expression profiling of Lgr5+ basal epithelial cells, we identify a new marker, Tetraspanin8 (Tspan8). Fractionation based on Tspan8 and Lgr5 expression uncovered three distinct mammary stem cell (MaSC) subsets in the adult mammary gland. These exist in a largely quiescent state but differ in their reconstituting ability, spatial localization, and their molecular and epigenetic signatures. Interestingly, the deeply quiescent MaSC subset (Lgr5+Tspan8hi) resides within the proximal region throughout life, and has a transcriptome strikingly similar to that of claudin-low tumours. Lgr5+Tspan8hi cells appear to originate from the embryonic mammary primordia before switching to a quiescent state postnatally but can be activated by ovarian hormones. Our findings reveal an unexpected degree of complexity within the adult MaSC compartment and identify a dormant subset poised for activation in response to physiological stimuli.


Subject(s)
Cell Cycle/drug effects , Hormones/pharmacology , Mammary Glands, Animal/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Movement , Cell Proliferation/drug effects , Female , Humans , Mammary Glands, Animal/drug effects , Mice , Stem Cells/drug effects , Stem Cells/metabolism
15.
Development ; 144(6): 1065-1071, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27993977

ABSTRACT

Advances in stem cell research have enabled the generation of 'mini organs' or organoids that recapitulate phenotypic traits of the original biological specimen. Although organoids have been demonstrated for multiple organ systems, there are more limited options for studying mouse mammary gland formation in vitro Here, we have built upon previously described culture assays to define culture conditions that enable the efficient generation of clonal organoid structures from single sorted basal mammary epithelial cells (MECs). Analysis of Confetti-reporter mice revealed the formation of uni-colored structures and thus the clonal nature of these organoids. High-resolution 3D imaging demonstrated that basal cell-derived complex organoids comprised an inner compartment of polarized luminal cells with milk-producing capacity and an outer network of elongated myoepithelial cells. Conversely, structures generated from luminal MECs rarely contained basal/myoepithelial cells. Moreover, flow cytometry and 3D microscopy of organoids generated from lineage-specific reporter mice established the bipotent capacity of basal cells and the restricted potential of luminal cells. In summary, we describe optimized in vitro conditions for the efficient generation of mouse mammary organoids that recapitulate features of mammary tissue architecture and function, and can be applied to understand tissue dynamics and cell-fate decisions.


Subject(s)
Mammary Glands, Animal/growth & development , Organoids/cytology , Tissue Culture Techniques/methods , Animals , Cell Lineage , Clone Cells , Epithelial Cells/cytology , Female , Flow Cytometry , Genes, Reporter , Imaging, Three-Dimensional , Mammary Glands, Animal/cytology , Mice , Microscopy, Confocal
16.
Methods Mol Biol ; 1501: 291-308, 2017.
Article in English | MEDLINE | ID: mdl-27796960

ABSTRACT

Lineage tracing analysis allows mammary epithelial cells to be tracked in their natural environment, thereby revealing cell fate and proliferation choices in the intact tissue. This technique is particularly informative for studying how stem cells build and maintain the mammary epithelium during development and pregnancy. Here we describe two experimental systems based on Cre/loxP technology (CreERT2/loxP and rtTA/tetO-Cre/loxP), which allow the inducible, permanent labeling of mammary epithelial cells following the administration of either tamoxifen or doxycycline.


Subject(s)
Cell Lineage/physiology , Mammary Glands, Animal/physiology , Stem Cells/physiology , Animals , Breast/physiology , Cell Lineage/drug effects , Doxycycline/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Female , Mammary Glands, Animal/drug effects , Mice , Stem Cells/drug effects , Tamoxifen/pharmacology
17.
Breast Cancer Res ; 18(1): 116, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27887631

ABSTRACT

Lineage tracing is increasingly being utilised to probe different cell types that exist within the mammary gland. Whilst this technique is powerful for tracking cells in vivo and dissecting the roles of different cellular subsets in development, homeostasis and oncogenesis, there are important caveats associated with lineage tracing strategies. Here we highlight key parameters of particular relevance for the mammary gland. These include tissue preparation for whole-mount imaging, whereby the inclusion of enzymatic digestion can drastically alter tissue architecture and cell morphology, and therefore should be avoided. Other factors include the scoring of clones in three dimensions versus two dimensions, the timing of induction, and the marked variability in labelling efficiency that is evident not only between different mouse models harbouring a similar gene promoter but also within a given strain and even within a single mammary gland. Thus, it becomes crucial to visualise extensive areas of ductal tissue and to consider the intricacies of the methodology for lineage tracing studies on normal mammary development and on potential 'cells of origin' of cancer.


Subject(s)
Cell Lineage , Mammary Glands, Animal/diagnostic imaging , Mammary Glands, Human/diagnostic imaging , Molecular Imaging , Animals , Biomarkers , Cell Lineage/genetics , Cell Tracking/methods , Clonal Evolution , Female , Humans , Imaging, Three-Dimensional/methods , Molecular Imaging/methods
18.
Cell Rep ; 16(1): 174-185, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27320914

ABSTRACT

Fas apoptotic signaling regulates diverse physiological processes. Acute activation of Fas signaling triggers massive apoptosis in liver. Upon Fas receptor stimulation, the BH3-only protein Bid is cleaved into the active form, tBid. Subsequent tBid recruitment to mitochondria, which is facilitated by its receptor MTCH2 at the outer mitochondrial membrane (OMM), is a critical step for commitment to apoptosis via the effector proteins Bax or Bak. MOAP-1 is a Bax-binding protein enriched at the OMM. Here, we show that MOAP-1-deficient mice are resistant to Fas-induced hepatocellular apoptosis and lethality. In the absence of MOAP-1, mitochondrial accumulation of tBid is markedly impaired. MOAP-1 binds to MTCH2, and this interaction appears necessary for MTCH2 to engage tBid. These findings reveal a role for MOAP-1 in Fas signaling in the liver by promoting MTCH2-mediated tBid recruitment to mitochondria.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , Liver/cytology , Liver/metabolism , Mitochondria/metabolism , fas Receptor/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/deficiency , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/deficiency , Fibroblasts/cytology , Fibroblasts/metabolism , HCT116 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Mice, Knockout , Mitochondrial Membrane Transport Proteins/metabolism , Protein Binding
19.
Nat Commun ; 7: 11400, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27102712

ABSTRACT

The mammary gland represents a unique tissue to study organogenesis as it predominantly develops in the post-natal animal and undergoes dramatic morphogenetic changes during puberty and the reproductive cycle. The physiological function of the mammary gland is to produce milk to sustain the newborn. Here we view the lactating gland through three-dimensional confocal imaging of intact tissue. We observed that the majority of secretory alveolar cells are binucleated. These cells first arise in very late pregnancy due to failure of cytokinesis and are larger than mononucleated cells. Augmented expression of Aurora kinase-A and Polo-like kinase-1 at the lactogenic switch likely mediates the formation of binucleated cells. Our findings demonstrate an important physiological role for polyploid mammary epithelial cells in lactation, and based on their presence in five different species, suggest that binucleated cells evolved to maximize milk production and promote the survival of offspring across all mammalian species.


Subject(s)
Aurora Kinase A/genetics , Cell Cycle Proteins/genetics , Epithelial Cells/metabolism , Lactation/physiology , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Animals , Aurora Kinase A/metabolism , Breast Feeding , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Nucleus/ultrastructure , Cell Size , Cytokinesis/genetics , Epithelial Cells/ultrastructure , Female , Gene Expression Regulation, Developmental , Humans , Mammary Glands, Animal/ultrastructure , Mammary Glands, Human/ultrastructure , Mice , Mice, Transgenic , Milk/metabolism , Milk/physiology , Pregnancy , Primary Cell Culture , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Polo-Like Kinase 1
20.
Nat Cell Biol ; 17(4): 365-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25730472

ABSTRACT

Expansion and remodelling of the mammary epithelium requires a tight balance between cellular proliferation, differentiation and death. To explore cell survival versus cell death decisions in this organ, we deleted the pro-survival gene Mcl-1 in the mammary epithelium. Mcl-1 was found to be essential at multiple developmental stages including morphogenesis in puberty and alveologenesis in pregnancy. Moreover, Mcl-1-deficient basal cells were virtually devoid of repopulating activity, suggesting that this gene is required for stem cell function. Profound upregulation of the Mcl-1 protein was evident in alveolar cells at the switch to lactation, and Mcl-1 deficiency impaired lactation. Interestingly, EGF was identified as one of the most highly upregulated genes on lactogenesis and inhibition of EGF or mTOR signalling markedly impaired lactation, with concomitant decreases in Mcl-1 and phosphorylated ribosomal protein S6. These data demonstrate that Mcl-1 is essential for mammopoiesis and identify EGF as a critical trigger of Mcl-1 translation to ensure survival of milk-producing alveolar cells.


Subject(s)
Epidermal Growth Factor/biosynthesis , Lactation/genetics , Lactation/metabolism , Mammary Glands, Animal/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Animals , Apoptosis/genetics , Base Sequence , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Cell Survival , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/metabolism , Female , Gene Knockout Techniques , Mice , Mice, Inbred C57BL , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Phosphorylation , Pregnancy , Ribosomal Protein S6/metabolism , Sequence Analysis, RNA , Stem Cells/cytology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...